IMMUNERESPONSEANDIMMUNOMODULATIONIN CHRONICHEPATITISBVIRUSINFECTION

D.SPRENGERS

Immune response and immunomodulation in chronic hepatitis B virus infection

Immuunresponsenimmunomodulatieinchronische HepatitisBvirusinfectie

PROEFSCHRIFT

terverkrijgingvandegraadvandoctor aandeErasmusUniversiteitRotterdam opgezagvandeRectorMagnificus Prof.dr.S.W.J.Lamberts envolgensbesluitvanhetCollegevoorPromoties.

Deopenbareverdedigingzalplaatsvindenop woensdag1november2006om13.45uur

door

Dave Sprengers

geborenteGoirle Promotiecommissie

Promotor: Prof.dr.H.L.A.Janssen

Copromotor: Dr.R.G.vanderMolen

Overigeleden: Prof.dr.J.H.P.Wilson Prof.dr.A.D.M.E.Osterhaus Prof.dr.S.W.Schalm

ThisstudywassupportedbyagrantfromTheNetherlandsOrganisationfor HealthResearchandDevelopment(ZonMw ,Agikostipendium dossiernum mer92003244).

Publicationofthisthesiswasfi nanciallysupportedby:ZonMw ,ZambonNederlandBV , NovartisPharmaBV, Aanmijnouders

TU REMANEBIS AMICUS MAXIMUS PERPETUO

Contents Page Chapter1: introduction 1

Chapter2: Functionalimpairmentofmyeloidandplasmacytoid 17 dendriticcellsofchronichepatitisBpatients

Chapter3: Flowcytometryoffi neneedleaspirationbiopsies:anew 33 methodtomonitorintrahepaticimmunologicalenvironmentin chronicviralhepatitis

Chapter4: Differentcompositionofintrahepaticlymphocytesinthe 45 immunetoleranceandimmuneclearancephaseofchronic hepatitisB

Chapter5: Analysisofintrahepatic HBVspecificcytotoxicTcellsduring 59 andafteracuteHBVinfectioninhumans

Chapter6: In vivo immunizationfollowingvirussuppression:anovel 73 approachforinducingimmunecontrolinchronichepatitisB

Chapter7: In vivo immunization incombinationwithpeginterferonfor 79 chronichepatitisBvirusinfection

Chapter8: InductionofC D4+CD25+regulatoryTcellsassociatedwith 93 nonresponsetopegylated Interferonαtherapyforchronic hepatitisBvirusinfection

Chapter9: Discussion 109

Summary 123 Samenvatting Dankwoord CurriculumVitae

Abbreviations

ALT alanineominotransferase antiHBc antibodiesagainsthepatitisBcoreantigen antiHBe antibodiesagainsthepatitisBeantigen antiHBs antibodiesagainsthepatitisBsurfaceantigen CHB chronichepatitisBvirus CTL cytotoxicTcells DC dendriticcells DNA deoxyribonucleicacid FNAB fin eneedleaspirationbiopsy GMCSF granulocyte/macrophagecolonystimulatingfactor HBcAg hepatitisBcoreantigen HBeAg hepatitisBeantigen HBsAg hepatitisBsurfaceantigen HBV hepatitisBvirus HCV hepatitisCvirus HDV hepatitisDvirus HIV humanimmunodeficiencyvirus HLA humanleukocyteantigen IFN interferon IL interleukin IVI invivoimmunization LIL liverinfiltratinglymphocytes mDC myeloiddendriticcells NKcell NaturalKillercell PB peripheralblood PBMC peripheralbloodmononuclearcells PCR polymerasechainreaction pDC plasmacytoiddendriticcells PegIFN pegylatedinterferon poly(I:C) polyriboinosinicpolyribocytidylicacid PreS1Ag hepatitisBpreS1antigen PreS2Ag hepatitisBpreS2antigen SAC Staphylococcus aureusCowenstrainI Th1/Th2 type1/type2Thelpercell Thcell Thelpercell TNF tumornecrosisfactor Treg regulatoryTcells

CHAPTER 1

Introduction

Modifiedfrom: ImmunomodulatorytherapyforchronichepatitisBvirusinfection D.Sprengers,H.L.A.Janssen,FundamClinPharmacol2005;19:1726

INTRODUCTION

Chronic hepatitis B virus infection HepatitisBvirus(HBV)isoneofthemostprevalentviralpathogensofmanwithalmost athirdoftheworldpopulationhavingevidenceofinfectionandaround350million chronicallyinfectedpatients 1.ChronichepatitisBischaracterizedbyinflammatory liverdiseaseofvariableseverityandisassociatedwithasignificantlyincreasedrisk ofdevelopingcirrhosis,liverfailureandhepatocellularcarcinoma1,2. HBVisanoncytopathicvirusandliverinjuryismainlymediatedbythehostimmune response against virus infected liver cells and by the production of inflammatory cytokines.Avigorous,polyclonalandmultispecificcytotoxic(CTL)andhelperT(Th) cellresponsetoHBVisreadilydetectableintheperipheralbloodofpatientswithacute selflimitedhepatitisB,butisweak,antigenicallyrestrictedorundetectableinpatients withchronicHBVinfection3.ThisTcellresponseisbelievedtoberesponsibleforthe eliminationofthehepatitisBvirus. Twomajorclassesofantiviraltherapeuticshavebeenadoptedtotreattheinfection: drugsthatdirectlyinterferewithvirusreplicationanddrugsthatmodulateantiviral immuneresponse.Inmostcountrieslamivudine,adefovirdipivoxilandentecavirare the only approved inhibitors of viral replication (HBVDNA polymerase inhibitors). Although lamivudine leads to rapid and almost absolute discontinuation of HBV replication4,breakthroughofresistantHBVvariantslimitslongtermuse5,6.These limitations underline the need for specific and nonspecific immunotherapeutic strategiesinordertoenhanceorbroadenthedefectiveTcellresponseinchronically infectedpatients. In this chapter we will focus on the immune response to HBV and we will review reported data on immunotherapeutic strategies like immunomodulatory drugs (cytokines andThymic derivates) and vaccine therapies using currently available recombinant antiHBV vaccines, lipopeptidebasedTcell vaccine and newly developedgeneticvaccines.

The hepatitis B virus HBVisapartlydoublestrandedDNAvirusbelongingtothegroupofhepadnaviridae 7(fig1).ThereplicationofHBVisbelievedtooccurpreferentiallyinthehepatocyte. Analysisofthenucleotidesequencerevealed4openreadingframes,regionsofthe genomewitchmaycodeforviralantigens7.AlthoughtheHBVgenomecontainsonly 3200nucleotides,itscompactnessandcircularcomposition,employingoverlapping for production of several viral antigens, make the virus highly efficient for replication.Afterentryintothehepatocyte’snucleusthevirusisuncoatedandthe genomicDNAisconvertedtoasupercoiledformofcovalentlyclosedfullydouble strandedDNA,whichistranscribedtopregenomicmessengerRNA(mRNA).This viralmRNAistransportedtothecytoplasmwhereitcodesfortheproductionofviral .mRNAisreversetranscribedintoaminusstrandofDNAwhichisutilized asatemplateforcompletionoftheplusstrandofthenewlysynthesizedviralDNA. The HBVDNApolymerase exerts both aDNA polymerase activity and a reverse transcriptaseactivity.OutoftheseviralproteinsandviralDNAinthecytoplasmofthe

3

hepatocytenewHBVparticlesareassembled,thatsubsequentlyleavethecellinto thecirculation.

HBsAg

HBcAg

HBV polymerase

HBV DNA (double stranded)

Figure1.SchematicoverviewofanHBVparticle,alsoknownastheDaneparticle. TheHBVgenomeencodesforseveralproteinsthatareusedtoassembletheDane particle.TheoutersurfaceofthevirusconsistsofthreetypesofhepatitisBsurface antigen(HBsAg).HepatitisBcoreantigen(HBcAg)formstheinnercoreoftheDane particle.TheviralDNAandtheHBVpolymeraseresideinsidetheinnercoreofthe virus.

The immune response to the hepatitis B virus in humans Patients infected with HBV may develop one of two types of antiHBV immune responses.Thefirstisaneffectiveantiviralresponsethatsuppressesviralgrowthas theresultofbothnonspecific(innate)aswellasspecific(adaptive)immunity.After earlyactionsofnaturalkiller(NK)cells,NKTcellsandantiviralcytokines,individuals withacuteselflimitedHBVinfectionmountavigorouspolyclonalandmultispecific ThandCTLresponsetoepitopeswithintheHBVenvelope(HBe),nucleocapsidand polymeraseproteins.ThepeptidesarepresentedtotheTcellsbyprofessionalantigen presentingcells(APC)andonthesurfaceofinfectedhepatocytesinthecontextofa MHCclassImolecule(fig2).Theresponseisreadilydetectableinperipheralblood 811.It often coincides with an elevation of serum alanineaminotransferase levels andprecedesclearanceofHBeandHBs(surface)antigensandthedevelopmentof neutralizingantibodies.UponrecognitionofviralpeptidesCTLacquirethecapacity toeithercureHBVinfectedcellsviaanoncytopathic,cytokinemediatedinhibition ofHBVreplication,ortokillthemviaperforinFasligandandTNFαmediateddeath pathways1215.Botheffectorfunctionshavebeenobservedduringresolutionofacute hepatitisBandthistype1Tcell(Th1)responsepersistsevenafterclinicalrecovery16. Incontrast,thesecondtypeofantiHBVimmuneresponseisanineffectiveone.The HBVspecificimmuneresponseisweak,antigenicallyrestrictedorundetectableinthe bloodofchronicallyinfectedpatients,althoughindividualHBVspecificTcellclones havebeenisolatedandexpandedfromliverbiopsies17,18.SinceHBVisconsidereda noncytopathicvirusandthedegreeofintrahepaticinflammatoryleukocyticinfiltrateis regardedasthehistologicalhallmarkoftheseverityofchronichepatitisB,ithasbeen

4 INTRODUCTION postulatedthattheHBVspecificimmuneresponseistooweaktoeliminateHBVfrom allinfectedhepatocytes,butsufficientlystrongtocontinuouslydestroyHBVinfected hepatocytesandtoinducechronicinflammatoryliverdiseaseinpersistentlyinfected individuals.Thereasonforthisinefficient,yetharmfulnatureofthecellularimmune responseinchronichepatitisBiscurrentlynotknown(table1),butitemphasizesthe needforimmunomodulatorydrugscapableofinducinganeffectiveTcellresponse thatenableseradicationofchronicHBVinfection.

Infected hepatocytes IFN-α prevents infection of Cytotoxic neighboring cells T cell Killing infected cell (specific)

HBV-particles T cell

Cell killing ADCC (nonspecific)

Antigen T helper NK cell Presenting Cell cell

Soluble factors regulate immune response (cytokines)

Antibody B cell producing cell

Figure2.SchematicoverviewofcellularimmuneresponsetothehepatitisBvirus. ViralantigensareprocessedandpresentedtoTcells(ontheinfectedcellsurface) andBcells(freeantigenicpiecesorviruses)byAntigenPresentingCells.Complexing ofantibodyproducedbyBcells,withvirus(opsonization)increasestheengulfment ofthevirusbyphagocytesandcanneutralisevirus.Bindingofantibodytoinfected targetcellsactivatesantibodydependentcellmediatedcytotoxicity(ADCC).Tcells (helperaswellascytotoxic)producecytokineswithnumerousimmunoregulatory actions.ProductsofTcellsactivatemacrophagesforkillingofingestedvirusand naturalkillercellsfornonspecificcytotoxicityagainstvirusinfectedcells.Interferons (IFNα/β)areproducedbyinfectedhepatocytesforprotectionofsurroundingcells againstvirusinfection.

5

Treatment of chronic hepatitis B TheaimoftreatmentofchronicviralhepatitisBistosuppressHBVreplicationbefore significant irreversible liver damage occurs.Ideally, the primary goals of therapy aretosuppressviralreplicationandtoinduceremissionofliverdisease.Thelong term goals of treatment are to eliminate the virus, prevent disease progression to cirrhosis or liver cancer and to improve patient survival.Definitions of responses to antiviral therapy can be classified as virologic, biochemical and histological 19. InpatientswhowereinitiallyHBeAgpositiveeffectivetreatmentofchronicHBVis definedassustainedclearanceofcirculatingHBeAgwithproductionofantibodiesto HBeanddecreaseofHBVDNAto<105geq/ml(virologicresponse);improvement in liver disease as determined by normalisation of serum alanine transaminase levels(biochemicalresponse)andreductionofnecroinflammationonliverbiopsies (histological response).In active HBeAg negative chronic hepatitis, characterized bydetectionofantiHBeantibodieswithoutHBeAginserum,detectableHBVDNA usingnonPCRbasedmethods,elevatedalaninetransaminaselevelsandhepatic necroinflammation 20,virologicresponseisdefinedasdecreaseofHBVDNAto< 105geq/ml19. Reportsonimmunomodulatoryantiviralagentsthatareunderreview(table2)willbe analysedfortheseresponseparameters.

Table 1. Possible causes of persistent HBV infection

Factor Viral factors - High viral load - High replication rate - Viral inhibition of antigen presentation - Viral mutations that abrogate, anergize or antagonize antigen recognition by specific T-cells - Immunosuppressive effects of virus

Host factors - Immunological tolerance - Exhaustion of T-cell response - Insufficient co-stimulation of virus-specific T-cells - Infection of immunologically privileged sites - Host HLA background - Inefficient viral antigen processing by antigen presenting cells or hepatocytes - Inefficient responsiveness of B- or T-cells; alteration of the Th1-Th2 balance Immunotherapeutic strategies to control chronic hepatitis B virus infection cytokines Interferon-α therapy Interferons are cytokines with immunomodulatory, antiproliferative and antiviral properties.Interferonα(IFNα)hasmarkedimmunoregulatorybutlesspronounced antiviraleffects21,22.IFNαwaslicensedin1992andafterbeingextensivelystudied,it isthefirstapprovedtreatmentforchronicHBVinfectioninmostcountries.IFNαhas beenshowntoinhibitviralRNApregenomicpackagingintocoreparticlesandalsoto

6 INTRODUCTION

enhanceexpressionofHBsAgonhepatocytes23. Multiple studies ofIFNα therapy in chronic HBV have demonstrated beneficial effects.Asustainedresponseimprovessurvival,butfrequencyofresponseislow 2426.Accordingtoametaanalysiswhichincluded15randomisedcontrolledstudies involving 837 patientsIFNα achieves HBeAgseroconversion in 33% of treated patientsusing16weeksoftherapycomparedto12%ofcontrols.Importantly,loss ofHBsAgwasrecordedin7.8%ofIFNtreatedpatientscomparedwithonly1.8%of controls.LossofdetectableHBVDNA,asdeterminedbyhybridisationassay,and normalisationofALTlevelwerealsomorecommonintreatedthancontrolpatients 27.However,resultswereobtainedinselectedpatientcohortswithmarkeddisease activityatbaseline.Recentlargestudiesshowedresponseratesof20% 28.There was a strong correlation between baseline ALT and HBeAg seroconversion rate: approximately510%forserumALTtwofoldelevated,20%forbaselineALToftwo fivefold the upper limit of normal and 3040% forALT fivefold elevated. Post hoc analysis also revealed other major pretreatment factors correlated with response toIFNα:lowHBVDNAlevelsandincreasedscoresofactivityandfibrosisonliver biopsy19. The optimal duration of interferon therapy for HBV is not well established.In a large multicenter trial it was demonstrated that continuing therapy for 32 weeks was beneficial in patients who did not clear HBeAg after 16 weeks, but who had atthattimelowlevelsofHBVDNA(<10pg/ml) 29.Thedurabilityofresponsesand continued benefit ofIFNα therapy were recently assessed in several longterm followupstudies.StudiesfromNorthAmericaandEuropereportedthat95100%of respondersremainedHBeAgnegativeduring510yearsoffollowupandultimately 3086%ofresponderslostHBsAg.Incontrast,Asianstudiesshowonlylowrateof durableresponsewithnoobviousdifferencebetweencontrolsandtreatedpatients 3033. PatientswhowereinitiallyHBeAgnegativeappeartorelapsemoreoftenafterIFN α therapy than HBeAg positive patients.In HBeAgnegative chronic viral hepatitis responseratesarehighlyvariablewith624%ofpatientsmaintainingasustained response1218monthsaftercessationoftherapy34,35.Morerecentlyastudyshowed abeneficialeffectof24monthsofIFNαtherapy,with30%sustainedrespondersina selectedgroupofHBeAgnegativepatients30.

Recently, PegylatedIFNα (PegIFN) was developed through the process of pegylation,inwhichapolyethyleneglycol(PEG)polymermoleculeisattachedtothe baseIFNαmoleculetoproduceadrugwithaprolongedhalflife 36.Aftershowing considerableimprovementinefficacyoverconventionalIFNαinchronichepatitisC infection,preliminaryresultsshowedthataftera24weeksfollowuptwiceasmany HBeAgpositivepatientsreceivingPegIFNα2afor6monthsachievedbothvirologic andbiochemicalresponsecomparedwithpatientsreceivingconventionalIFNα;24% vs,12%respectively37.Inalargemulticentre,doubleblindcontrolledtrialPegIFNα 2bmonotherapyfor52weeksinducedlossofHBeAgin36%ofpatientsafter26

7

weeksoffollowup38.TheseresultsindicatethatPegIFNtreatmentisatleastequal butprobablysuperiorinefficacytoconventionalIFNαforHBeAgpositiveCHB. Additionally, in this multicentre trial it was investigated whether a combination of PegIFNα2bandthenucleosideanaloguelamivudinehasbeneficialeffects.The combinationPegIFNα2b–lamivudineresultedinahigherHBeAgseroconversion rateatendoftreatmentcomparedtopatientstreatedwithPegIFNα2bmonotherapy, butequalresponseratesattheendoffollowup.Resultsofalargestudy(n=537) comparing 48 weeks of PegIFNα2a with and without lamivudine to lamivudine alone, in the treatment of HBeAgnegative patients, showed that the combination ofPegIFNα2awithlamivudinedidnotimproveresponseratesoverPegIFNα2a monotherapy.Attheendof24weeksfollowupvirologicresponsewasachievedin 43%PegIFNα2atreatedpatientsvs.44%ofpatientstreatedwiththecombination therapy39.

Table 2. Immunological functions of components used as immunomodulatory drugs to improve the immune response to HBV Component IFN-α - Inhibits viral replication; degrades viral components - Induces production of IL-15 and consequently enhances T- cell growth - Augments lytic activity of NK cells and CTL - Enhances expression of cell-surface antigens by the major histocompatibility complex (MHC) - Modulates production of proinflammatory cytokines like IL-1, TNF-α and IL-8 IL-2 - Activates T cells and induces differentiation of B cells; growth factor of T- and B-cells - Stimulates production of antiviral cytokines TNF-α and IFN-γ IL-12 - Proliferation of pre-activated T-cells and NK cells - Enhances generation of CTL - Augments cytotoxic activity of CTL and NK cells - Induces production of antiviral cytokines TNF-α and particularly IFN-γ GM-CSF - Stimulates differentiation of hematopoietic precursor cells - Major stimulatory cytokine for viability, differentiation and function of DC, the most important cell type for stimulation of primary T-cell mediated immune responses. - Increases expression of MHC class II molecules on antigen presenting cells. Thymosine-α - Stimulates activity of NK cell-mediated cytotoxicity - Stimulates production of IL-2 and IFN-γ - Increases maturation of T-cells Vitamin E - Increases lymphocyte proliferation and IL-2 production - Raises activity of NK cells

Interleukine-2 Interleukine2(IL2)hasbeenknownformanyyearsasaTcellgrowthpromoting factor.IthasbeendemonstratedinapilotstudythatIL2enhancesCD4+Tcellsand NKcellsinchronichepatitisB40.DespitethereportofclearanceofHBsAgandthe appearanceofantiHBsantibodiesinanHIVinfectedpatientafterrecombinantIL2 therapy 41,apilotstudyofnaturalhumanIL2inpatientswithchronicHBVdidnot

8 INTRODUCTION

resultineitherabiochemicalorimmunologicalresponse42.Arandomisedcontrolled trialofIL2versusplaceboinHBVpatientscoinfectedwithHIVdidshowasignificant increaseinCD4+TcellcountbutIL2treatmentdidnotmodifyHBVreplication43. Inadoubleblindrandomisedcontrolledtrial31patientswithchronicHBVinfection receiveddifferentdosesofIL2subcutaneously44.Nodifferencesinthefrequencyof lossofHBeAgwereobservedbetweentreatedandplacebogroupsanddecreasein serumHBVDNAconcentrationwassimilarinbothpopulations.Studiesinvestigating IL2 in combination with other antiviral therapies have also been performed. A randomised controlled trial showed no additional effect of a combination ofIL 2 withIFNα, with respect to HBVDNA decrease, HBeAg seroconversion or ALT normalisation 45.Combining HBsAg vaccination with lamivudine only or with lamivudineandIL2didnotshowanyadditionalefficacyofIL246.

Interleukine-12 Interleukine12(IL12)isaproductofactivatedinflammatorycellslikemonocytes, macrophages and dendritic cells 47.IL12 has a direct proliferative effect on pre activatedTcellsandNKcells,anditinducesthesecellstoproducecytokines,such asgranulocytemacrophagecolonystimulatoryfactor,TNFαandparticularlyIFNγ. Therefore,IL12favoursthedifferentiationofTh1cells. ItwasshownthatamongstpatientswithchronicHBVundergoingIFNαtreatment, onlythosewhoclearHBVshowasubstantialincreaseintheproductionofbiologically activeIL1248.ThepeakofserumIL12occurredafterthehepatitisflarebutpreceded orcoincidedwithHBeAgseroconversion.Recentlyamulticentreopenlabelphase I/IItrialwasconductedtotesttheefficacyofonceweeklyinjectionsof0.03,0.25and 0.50g/kgrHuIL12overa12weekcourse49.rHuIL12wasgenerallywelltolerated, althoughassociatedwithtransientdecreasesofneutrophilsandlymphocytecounts, andwithelevationsinserumtransaminasesandbilirubin.ALTnormalisationtogether withsustainedclearanceofHBVDNAandHBeAgwasobservedattheendoffollow upin3/46patients,oneineverydosegroup.TwopatientsseroconvertedtoantiHBe. Thus,theantiviralactivityofrHuIL12inpatientswithchronicHBVdoesnotappear to be advantageous compared to other currently available treatments.Alternative dosingstrategiesmayincreasetheantiviralefficacyofIL12.

Granulocyte-macrophage colony-stimulating factor Recombinanthumangranulocytemacrophagecolonystimulatingfactor(rhGMCSF) isthemajorstimulatorycytokinefortheviability,differentiationandfunctionofdendritic cells50.ItaugmentstheexpressionofTcellcostimulatorymoleculesandincreases theexpressionofMHCclassIImolecules,whicharecrucialforantigenrecognition andinitiationofanimmuneresponsebyCD4+Tcells5153.Theconsequencesofthe immunostimulatory activity of rhGMCSF have been enhancedTcell proliferative responses to suboptimal antigen concentrations and the augmentation of primary antibodyresponsesin vitroandin vivo54. Apilotstudyincluding9patientswithchronicHBVinfectionshowedthatrhGMCSF

9

therapy for 6 weeks significantly enhancedTNFα andIL1 production in cultured mononuclear cells. A significant reduction of serum HBVDNA levels was also observed55.InapreliminarystudyforpreviousIFNnonrespondersrhGMCSFplus IFNα for 4 months led to HBVDNA negativity by a hybridization assay in 5 of 8 patients56.ThesedatasuggestthatthecombinationofrhGMCSFandIFNαmaybe moreeffectiveatclearingHBVDNAthanIFNαalone.

Vaccine therapy for chronic hepatitis B virus infection Immunotherapy using recombinant anti-HBV vaccine. PilotclinicalstudiesestablishedthatspecificvaccinetherapybystandardantiHBV vaccination(GenHevacB)couldcancelorreduceHBVreplicationinofchroniccarrier subjects57,58.Theseresultswereconfirmedinarecentcontrolledstudy59showing boththeefficacyandlimitationsof(standard)vaccinetherapyinchronicinfection. The118includedpatientswere‘naive’subjectswhohadneverreceivedanyprevious HBVtherapy.Theyweregiveneitherfiveintramuscularinjectionsof20gofapreS2/ Svaccine(GenHevacB,PasteurMerieux;n=46),anSvaccine(RecombivaxMerck &Co.;n=34)orplaceboasacontrol(n=37).After12monthsfollowupandfive vaccineinjections,therewasnodifferenceintherateofserumHBVDNAreduction as determined by a hybridisation assay, between vaccinated and unvaccinated subjects.However,inthefirst6monthsfollowingvaccinationpatientstreatedwith eithervaccineweresignificantlymorelikelytoclearserumHBVDNAandseroconvert toantiHBethanuntreatedcontrols.At12monthsthesedifferenceslostsignificance due to increased seroconversion in the placebo group.Disappearance of serum HBsAg was not observed.In another randomized placebo controlled therapeutic vaccinationtrialincluding22chronicallyinfectedpatients,avaccinecontainingpre S1,preS2andSantigeniccomponents,didnotinduceanHBsAgspecificinduction ofTh1lymphokinesorHBVspecificCD8+Tcells 60.Thisresultmightexplainwhy atherapeuticsurfacevaccine,althoughsafeandwelltoleratedismostlynotableto breaktoleranceleadingtotheclearanceofthehepatitisBvirus.

T cell vaccine for chronic hepatitis B virus infection. Thegoalofanalternativevaccinationstrategyistousepeptideepitopesthatare recognizedbycytotoxicTlymphocytes(CTL)asimmunogensforthedevelopmentof prophylacticandtherapeuticvaccines.Recently,alipopeptidevaccine(CY1899)was designedconsistingofHBVcoreantigenpeptide1827astheCTLepitope,tetanus toxoidpeptide830843astheThelperpeptide,andtwopalmiticacidmoleculesas thelipids.Adoseescalationtrial(5,50,and500g)carriedoutin26normalsubjects showedthatthisvaccinewassafeandabletoinduceaprimaryHBVspecificCTL response61.Theaimofafollowingstudywastodeterminewhetherrepeateddoses ofCY1899givensubcutaneouslyto19patientswithchronichepatitisBcouldinitiate invivoCTLactivityandviralclearance62.Patientsreceivedupto4doses(ranging from0.05mgto15mg)6weeksapart.Administrationofthesingleepitopevaccine, CY1899,initiatedCTLactivity,butofamagnitudelowerthanthatobservedduring

10 INTRODUCTION

spontaneous HBV clearance.This lowlevelCTL activity was not associated with viraldeclineandnosignificantchangesinliverbiochemistryorviralserologywere observedduringfollowup.

DNA vaccines for chronic hepatitis B virus infections NewapproachesofvaccinationforHBVarenowbasedonintramuscularinjection of plasmidDNA encoding HBV antigen 63.The socalledDNA vaccines induce immuneresponsesagainstantigenssynthesizedin vivoafterintroductionofDNA’s encodingantigensequences.InthecontextofclassIandclassIIMHCmolecules, theendogenoussynthesisofantigenleadstoappropriateantigenpresentationwhich willresultinstronghumoralandcellularimmuneresponses.Thisnovelapproachto immunizationmayovercomefailuresofthetraditionalantigenbasedapproachand mayprovideeffectivetherapeuticvaccines. Afterbeingextensivelystudiedinanimalmodels63,64,recentlyinaphaseIclinicaltrial threegroupsofhealthyvolunteers(n=4eachgroup)receivedthreeadministrations ofDNA encoding the surface antigen of HBV at a dose of 1, 2, or 4 g 65.The vaccinewassafeandwelltolerated.Allthevolunteersdevelopedprotectiveantibody responses.InvolunteerswhowerepositivefortheHLAclassIA2allele,thevaccine

alsoinducedantigenspecificCD8+TcellsthatboundHLAA2/HBsAg335–343tetramers, secretedIFNγandlysedtargetcellspresentinganHBsAgCTLepitope.Enumeration of HBsAgspecificTcells producing cytokines indicated preferential induction of a Type 1Thelper cell response.These results provide the first demonstration of a DNAvaccineinducingprotectiveantibodytitresandbothhumoralandcellmediated immuneresponsesinhumans.Furtherstudiesareneededtoevaluatetherapeutic efficacyofDNAvaccinesinthesettingofchronicHBVinfection.

Other immunomodulators Thymosine alpha1 Thymosinealpha1isabovinethymusextractthatacceleratesthereplenishmentand maturationofthymocytes,stimulatesdifferentiationintoactiveTcellsandrestores TcellfunctionbyTcellmediatedantibodyproduction66.Thymosinealpha1(Tα1) concentrationsarelowinpatientswithchronicHBV 67.TrialsofTα1suggestthat itmaybeatleastaseffectiveasIFNαinpatientswithraisedserumALTwhoare HBeAgandHBVDNApositive68,69.Arecentmetaanalysiscontaining353patients evaluated the efficacy ofTα1 68, 7072.Tolerance toTα1 was good in all trials. Comparedtoplacebotherewasnosignificantbiochemicalresponsebutoddsratio forvirologicresponsetoTα1,measuredbyHBeAgseroconversionrateandHBV DNAclearanceinahybridizationassay,attheendoftherapy,at6andat12months posttreatmentwere0.56,1.67and2.67respectively.Finallytherewasasignificant increaseinvirologicresponseovertimeafterdiscontinuationofTα1treatment.This suggeststhatviralclearancemaybenoncytoliticandmaybemediatedbyIFNγand TNFαsecretedbyactivatedCTL73.ItwasshownthatIFNγproducingCD4+Tcells areincreasedinperipheralbloodofTα1treatedpatientsandthatNKTcellsandCTL

11

appeartobeaugmentedintheliver74. Fewstudieshaveconcentratedoncombinationtherapy.Arecenttrialanalysedthe efficacyofTα1plusfamciclovirfor26weeksvs.famcicloviralonevs.placebo75.The combinationgroupshowedsignificantlyhigherHBVDNAreduction, andat52weeks 5patients(15.6%)experiencedHBeAgseroconversionvs.noneintheothergroups. SustainedserologicalclearanceofHBeAgwasassociatedwithactivationofCD4+ HBVspecificTcellreactivity. Vtamin E VitaminE(αtocopherol)isanessentialvitaminwithantioxidantproperties 76that isalsoabletoenhancethecellmediatedimmunity76,77.Recentstudiesshowedits possible beneficial role in the treatment of chronic hepatitisCvirus infection 78, 79. Inarandomisedcontrolledpilotstudy15–ofwhich12HBeAgnegativepatients withchronicHBVinfectionweretreatedforthreemonthswith 300 mgtwicedaily 80. Baseline ALT and HBVDNA were comparable to 17 nontreated patients. At theendofafollowupof15monthsaftertherapy,ALTandHBVDNAserumlevels weresignificantlylowerinpatientsreceivingvitaminE,with7subjectsexhibitinga completeresponse(ALTnormalandHBVDNAnegativeinhybridisationassay)vs. noneofthecontrols. Vitamin E may probably be used as a supplementary treatment rather than monotherapyforchronicHBVinfection.Largerstudiesareneededtoconfirmthese interestingresultsandtoevaluatedifferentdosingregimesandcombinationswith otherantiviraldrugs.

Conclusions The primary aim in the treatment of chronic hepatitis B is to induce sustained disease remission and prevent liver failure and/or hepatocellular carcinoma.The recentemergenceofdrugresistantHBVmutantsandposttreatmentrelapseasa consequence of nucleoside analogue monotherapy emphasizes that the principal goalshouldbetostimulateasuccessfulimmuneresponse. AtpresenttheonlyevidenteffectiveimmunomodulatorytherapyisIFNα.Compared toconventionalIFNαPegIFNappearstoachievehigherresponseratesinHBeAg positivepatients.BothinHBeAgpositiveandHBeAgnegativepatientscombination of PegIFN and lamivudine was not superior to PegIFN alone.Important antiviral and immunomodulatory effects of other cytokines in combination with pegylated interferonα,maybemoresuccessfulthanmonotherapywitheithercomponent. Future immunotherapy approaches should aim to induce a coordinate activation of all components of the adaptive immune response to restore a multispecific antiHBVimmuneresponse.VaccinesabletoelicitbothHBVspecificTandBcell responsesinhumans,againstmultipleHBVantigens,presentalogicalandpromising immunomodulatorystrategy.However,manyfactors,likeantigenselection,frequency andtimingofvaccinationandsafetyissues,needtobeevaluatedbeforetherapeutic vaccination can be applied to induce durable disease remission in patients with chronicHBVinfection.

12 INTRODUCTION

References 1. KaneM.GlobalprogrammeforcontrolofhepatitisBinfection.Vaccine1995;13Suppl1:S479. 2. WrightTL,LauJY.ClinicalaspectsofhepatitisBvirusinfection.Lancet1993;342:13404. 3. ChisariFV,FerrariC.HepatitisBvirusimmunopathogenesis.AnnuRevImmunol1995;13:2960. 4. BenhamouY,DohinE,LunelFabianiF,etal.EfficacyoflamivudineonreplicationofhepatitisBvirusin HIVinfectedpatients.Lancet1995;345:3967. 5. LaiCL,ChienRN,LeungNW,etal.AoneyeartrialoflamivudineforchronichepatitisB.AsiaHepatitis LamivudineStudyGroup.NEnglJMed1998;339:618. 6. AngusP,VaughanR,XiongS,etal.Resistancetoadefovirdipivoxiltherapyassociatedwiththeselection ofanovelmutationintheHBVpolymerase.Gastroenterology2003;125:2927. 7. TiollaisP,PourcelC,DejeanA.ThehepatitisBvirus.Nature1985;317:48995. 8. MainiMK,BoniC,LeeCK,etal.TheroleofvirusspecificCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 9. RehermannB,LauD,HoofnagleJH,ChisariFV.CytotoxicTlymphocyteresponsivenessafterresolutionof chronichepatitisBvirusinfection.JClinInvest1996;97:165565. 10. LivingstonBD,CrimiC,GreyH,etal.ThehepatitisBvirusspecificCTLresponsesinducedinhumansby lipopeptidevaccinationarecomparabletothoseelicitedbyacuteviralinfection.JImmunol1997;159:1383 92. 11. GuidottiLG,AndoK,HobbsMV,etal.CytotoxicTlymphocytesinhibithepatitisBvirusexpressionby anoncytolyticmechanismintransgenicmice.ProcNatlAcadSciUSA1994;91:37648. 12. RehermannB.IntrahepaticTcellsinhepatitisB:viralcontrolversuslivercellinjury.JExpMed2000;191: 12638. 13. GanemD. Persistent infection of humans with hepatitis B virus: mechanisms and consequences.Rev InfectDis1982;4:102647. 14. GuidottiLG,RochfordR,ChungJ,ShapiroM,PurcellR,ChisariFV.Viralclearancewithoutdestructionof infectedcellsduringacuteHBVinfection.Science1999;284:8259. 15. HeiseT,GuidottiLG,CavanaughVJ,ChisariFV.HepatitisBvirusRNAbindingproteinsassociatedwith cytokineinducedclearanceofviralRNAfromtheliveroftransgenicmice.JVirol1999;73:47481. 16. GuidottiLG,ChisariFV.Cytokinemediatedcontrolofviralinfections.Virology2000;273:2217. 17. Barnaba V, FrancoA,AlbertiA, BalsanoC, BenvenutoR, Balsano F.Recognition of hepatitis B virus envelopeproteinsbyliverinfiltratingTlymphocytesinchronicHBVinfection.JImmunol1989;143:2650 5. 18. FerrariC,PennaA,GiubertiT,etal.Intrahepatic,nucleocapsidantigenspecificTcellsinchronicactive hepatitisB.JImmunol1987;139:20508. 19. Lok AS, Heathcote EJ, Hoofnagle JH. Management of hepatitis B: 2000summary of a workshop. Gastroenterology2001;120:182853. 20. HadziyannisSJ,VassilopoulosD.HepatitisBeantigennegativechronichepatitisB.Hepatology2001;34: 61724. 21. OritaniK,KincadePW,ZhangC,TomiyamaY,MatsuzawaY.TypeIinterferonsandlimitin:acomparison ofstructures,receptors,andfunctions.CytokineGrowthFactorRev2001;12:33748. 22. LevyDE.Whenceinterferon?Varietyintheproductionofinterferoninresponsetoviralinfection.JExpMed 2002;195:F158. 23. Lau JY, Bain VG,NaoumovNV, Smith HM,Alexander GJ, WilliamsR. Effect of interferongamma on hepatitisBviralantigenexpressioninprimaryhepatocyteculture.Hepatology1991;14:9759. 24. NiederauC, HeintgesT, Lange S, et al. Longterm followup of HBeAgpositive patients treated with interferonalfaforchronichepatitisB.NEnglJMed1996;334:14227. 25. VilleneuveJP,CondreayLD,WillemsB,etal.Lamivudinetreatmentfordecompensatedcirrhosisresulting fromchronichepatitisB.Hepatology2000;31:20710. 26. van Zonneveld M, Honkoop P, Hansen BE, et al. Longterm followup of alphainterferon treatment of patientswithchronichepatitisB.Hepatology2004;39:80410. 27. WongDK,Cheung AM,O’Rourke K,NaylorCD,Detsky AS, Heathcote J. Effect of alphainterferon treatmentinpatientswithhepatitisBeantigenpositivechronichepatitisB.Ametaanalysis.AnnIntern Med1993;119:31223. 28. SchalmSW,HeathcoteJ,CianciaraJ,etal.Lamivudineandalphainterferoncombinationtreatmentof patientswithchronichepatitisBinfection:arandomisedtrial.Gut2000;46:5628. 29. JanssenHL,GerkenG,CarrenoV,etal.InterferonalfaforchronichepatitisBinfection:increasedefficacy ofprolongedtreatment.TheEuropeanConcertedActiononViralHepatitis(EUROHEP).Hepatology1999; 30:23843. 30. Lampertico P,DelNinno E, Vigano M, et al. Longterm suppression of hepatitis B e antigennegative

13

chronichepatitisBby24monthinterferontherapy.Hepatology2003;37:75663. 31. LokAS,Chung HT, Liu VW, MaOC. Longterm followup of chronic hepatitis B patients treated with interferonalfa.Gastroenterology1993;105:18338. 32. LinSM,SheenIS,ChienRN,ChuCM,LiawYF.Longtermbeneficialeffectofinterferontherapyinpatients withchronichepatitisBvirusinfection.Hepatology1999;29:9715. 33. Ikeda K, Saitoh S, Suzuki Y, et al.Interferon decreases hepatocellular carcinogenesis in patients with cirrhosiscausedbythehepatitisBvirus:apilotstudy.Cancer1998;82:82735. 34. ManesisEK,HadziyannisSJ.InterferonalphatreatmentandretreatmentofhepatitisBeantigennegative chronichepatitisB.Gastroenterology2001;121:1019. 35. Oliveri F, SantantonioT, Bellati G, et al. Long term response to therapy of chronic antiHBepositive hepatitisBispoorindependentoftypeandscheduleofinterferon.AmJGastroenterol1999;94:136672. 36. Kozlowski A, Harris JM.Improvements in PEGylation: pegylated interferons for treatment of hepatitisC.JControlRelease2001;72:21724. 37. CooksleyWG,PiratvisuthT,LeeSD,etal.Peginterferonalpha2a(40kDa):anadvanceinthetreatment ofhepatitisBeantigenpositivechronichepatitisB.JViralHepat2003;10:298305. 38. Janssen HLA, Senturk H, Zeuzem S, et al. Peginterferon alfa2B and lamivudine combination therapy comparedwithpeginterferonalfa2BforchronicHBeAgpositivehepatitisB:arandomizedcontrolledtrial in307patients.Hepatology2003;38:1323. 39. Marcellin P, Lau GKK, Bonino F, et al. Peginterferon alfa2a (40KD) (pegasys) monotherapy is more effectivethanlamivudinemonotherapyinthetreatmentofHBeAgnegativechronichepatitisB:73week results from a phaseIII, partially doubleblind study of pegasys alone vs. pegasys plus lamivudine vs. lamivudine.JHepatol2004;40suppl1:95A. 40. OnjiM,KondohH,HoriikeN,etal.Effectofrecombinantinterleukin2onhepatitisBeantigenpositive chronichepatitis.Gut1987;28:164852. 41. GianottiN,UbertiFoppaC, Boeri E, et al. Hepatitis B surface antigen clearance and appearance of antibodiesagainsthepatitisBsurfaceantigenaftertreatmentwithrecombinantinterleukin2inahuman immunodeficiencyvirusinfectedpatient.Hepatology2000;32:140910. 42. TilgH,VogelW,TratkiewiczJ,etal.Pilotstudyofnaturalhumaninterleukin2inpatientswithchronic hepatitisB.Immunomodulatoryandantiviraleffects.JHepatol1993;19:25967. 43. ThibaultV,DelaugerreC,CalvezV,CostagliolaD,TubianaR,KatlamaC.Interleukin2treatmentdoes notmodifyhepatitisBorCreplicationinhumanimmunodeficiencyvirusinfectedpatients:resultsfroma randomizedcontroltrial.Hepatology2002;35:2389. 44. ArtilloS,PastoreG,AlbertiA,etal.Doubleblind,randomizedcontrolledtrialofinterleukin2treatmentof chronichepatitisB.JMedVirol1998;54:16772. 45. BruchHR,KornA,KleinH,etal.TreatmentofchronichepatitisBwithinterferonalpha2bandinterleukin2. JHepatol1993;17Suppl3:S525. 46. Dahmen A, HerzogHauff S, Bocher WO, Galle PR, Lohr HF.Clinical and immunological efficacy of intradermalvaccinepluslamivudinewithorwithoutinterleukin2inpatientswithchronichepatitisB.JMed Virol2002;66:45260. 47. TrinchieriG.Interleukin12andtheregulationofinnateresistanceandadaptiveimmunity.NatRevImmunol 2003;3:13346. 48. Rossol S, Marinos G,Carucci P, Singer MV, WilliamsR,NaoumovNV.Interleukin12 induction ofTh1 cytokinesisimportantforviralclearanceinchronichepatitisB.JClinInvest1997;99:302533. 49. ZeuzemS,HopfU,CarrenoV,etal.AphaseI/IIstudyofrecombinanthumaninterleukin12inpatientswith chronichepatitisC.Hepatology1999;29:12807. 50. MarkowiczS,EnglemanEG.Granulocytemacrophagecolonystimulatingfactorpromotesdifferentiation andsurvivalofhumanperipheralblooddendriticcellsinvitro.JClinInvest1990;85:95561. 51. Fischer HG, Frosch S,Reske K,ReskeKunz AB. Granulocytemacrophage colonystimulating factor activatesmacrophagesderivedfrombonemarrowculturestosynthesisofMHCclassIImoleculesandto augmentedantigenpresentationfunction.JImmunol1988;141:38828. 52. MorrisseyPJ,BresslerL,ParkLS,AlpertA,GillisS.Granulocytemacrophagecolonystimulatingfactor augmentstheprimaryantibodyresponsebyenhancingthefunctionofantigenpresentingcells.JImmunol 1987;139:11139. 53. GermainRN. MHCdependent antigen processing and peptide presentation: providing ligands forT lymphocyteactivation.Cell1994;76:28799. 54. LiehlE,HildebrandtJ,LamC,MayerP.Predictionoftheroleofgranulocytemacrophagecolonystimulating factorinanimalsandmanfrominvitroresults.EurJClinMicrobiolInfectDis1994;13Suppl2:S917. 55. MartinJ,QuirogaJA,BoschO,CarrenoV.Changesincytokineproductionduringtherapywithgranulocyte macrophagecolonystimulatingfactorinpatientswithchronichepatitisB.Hepatology1994;20:115661.

14 INTRODUCTION

56. VanThielDH,FriedlanderL,KaniaRJ,MolloyPJ,HassaneinT,FarukiH.Apreliminaryexperiencewith GMCSFplusinterferoninpatientswithHBVandHCVresistanttointerferontherapy.JViralHepat1997; 4Suppl1:1016. 57. WenYM,WuXH,HuDC,ZhangQP,GuoSQ.HepatitisBvaccineandantiHBscomplexasapproachfor vaccinetherapy.Lancet1995;345:15756. 58. Pol S,CouillinI, Michel ML, et al.Immunotherapy of chronic hepatitis B by anti HBV vaccine. Acta GastroenterolBelg1998;61:22833. 59. PolS,NalpasB,DrissF,etal.EfficacyandlimitationsofaspecificimmunotherapyinchronichepatitisB.J Hepatol2001;34:91721. 60. Jung MC, GrunerN, ZachovalR, et al.Immunological monitoring during therapeutic vaccination as a prerequisite for the design of new effective therapies: induction of a vaccinespecificCD4+Tcell proliferativeresponseinchronichepatitisBcarriers.Vaccine2002;20:3598612. 61. VitielloA,Ishioka G, Grey HM, et al.Development of a lipopeptidebased therapeutic vaccine to treat chronicHBVinfection.I.InductionofaprimarycytotoxicTlymphocyteresponseinhumans.JClinInvest 1995;95:3419. 62. HeathcoteJ,McHutchisonJ,LeeS,etal.ApilotstudyoftheCY1899Tcellvaccineinsubjectschronically infectedwithhepatitisBvirus.TheCY1899TCellVaccineStudyGroup.Hepatology1999;30:5316. 63. ThermetA,RollierC,ZoulimF,TrepoC,CovaL.ProgressinDNAvaccineforprophylaxisandtherapyof hepatitisB.Vaccine2003;21:65962. 64. DavisHL,McCluskieMJ,GerinJL,PurcellRH.DNAvaccineforhepatitisB:evidenceforimmunogenicity inchimpanzeesandcomparisonwithothervaccines.ProcNatlAcadSciUSA1996;93:72138. 65. RoyMJ,WuMS,BarrLJ,etal.InductionofantigenspecificCD8+Tcells,Thelpercells,andprotective levels of antibody in humans by particlemediated administration of a hepatitis B virusDNA vaccine. Vaccine2000;19:76478. 66. LiCL,ZhangT,SaibaraT,etal.Thymosinalpha1acceleratesrestorationofTcellmediatedneutralizing antibodyresponseinimmunocompromisedhosts.IntImmunopharmacol2002;2:3946. 67. Sherman KE, JonesCC, Goldstein AL,Naylor PH. Low thymosin alpha1 concentrations in patients chronicallyinfectedwiththehepatitisBvirus.ViralImmunol1991;4:1959. 68. ChienRN, LiawYF,ChenTC,YehCT, SheenIS. Efficacy of thymosin alpha1 in patients with chronic hepatitisB:arandomized,controlledtrial.Hepatology1998;27:13837. 69. Andreone P,CursaroC, Gramenzi A, et al. A randomized controlled trial of thymosinalpha1 versus interferonalfatreatmentinpatientswithhepatitisBeantigenantibodyandhepatitisBvirusDNApositive chronichepatitisB.Hepatology1996;24:7747. 70. ChanHL,TangJL,TamW,SungJJ.TheefficacyofthymosininthetreatmentofchronichepatitisBvirus infection:ametaanalysis.AlimentPharmacolTher2001;15:1899905. 71. MutchnickMG,LindsayKL,SchiffER,etal.Thymosinalpha1treatmentofchronichepatitisB:resultsofa phaseIIImulticentre,randomized,doubleblindandplacebocontrolledstudy.JViralHepat1999;6:397 403. 72. ZavagliaC, SeveriniR,TinelliC, et al.A randomized, controlled study of thymosinalpha1 therapy in patientswithantiHBe,HBVDNApositivechronichepatitisB.DigDisSci2000;45:6906. 73. GuidottiLG,IshikawaT,HobbsMV,MatzkeB,SchreiberR,ChisariFV.Intracellularinactivationofthe hepatitisBvirusbycytotoxicTlymphocytes.Immunity1996;4:2536. 74. SugaharaS,IchidaT,YamagiwaS,etal.Thymosinalpha1increasesintrahepaticNKTcellsandCTLsin patientswithchronichepatitisB.HepatolRes2002;24:346354. 75. Lau GK,Nanji A, Hou J, et al.Thymosinalpha1 and famciclovir combination therapy activatesTcell responseinpatientswithchronichepatitisBvirusinfectioninimmunetolerantphase.JViralHepat2002; 9:2807. 76. MeydaniM.VitaminE.Lancet1995;345:1705. 77. Gogu SR, Blumberg JB. Vitamin E increases interleukin2 dependent cellular growth and glycoprotein glycosylationinmurinecytotoxicTcellline.BiochemBiophysResCommun1993;193:8727. 78. vonHerbayA,StahlW,NiederauC,SiesH.VitaminEimprovestheaminotransferasestatusofpatients sufferingfromviralhepatitisC:arandomized,doubleblind,placebocontrolledstudy.FreeRadicRes1997; 27:599605. 79. OtaY,SasagawaT,SuzukiK,etal.VitaminEsupplementationincreasespolyunsaturatedfattyacidsof RBCmembraneinHCVinfectedpatients.Nutrition2004;20:35863. 80. Andreone P, Fiorino S,CursaroC, et al. Vitamin E as treatment for chronic hepatitis B: results of a randomizedcontrolledpilottrial.AntiviralRes2001;49:7581.

15

CHAPTER 2

Functional impairment of myeloid and plasmacytoid dendritic cells of chronic hepatitis B patients

R.G.vanderMolen, 1D.Sprengers,1R.S.Binda, 1E.C.deJong, 2H.G.M.Niesters 3 J.G.Kusters,1J.Kwekkeboom,1andH.L.A.Janssen1

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofCellBiologyandHistology,AcademicMedicalCenter,University ofAmsterdam,Amsterdam,TheNetherlands 3Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

Hepatology2004;40:738746 CHAPTER2

Abstract

Dendritic cells (DC) play an important role in the induction ofTcell responses. WehypothesizethatthehamperedantiviralTcellresponseinchronichepatitisB patientsisaresultofimpairedDCfunction.Inthisstudy,wecomparedthenumber, phenotypeandfunctionalityoftwoimportantbloodprecursorDC,myeloidDC(mDC) andplasmacytoidDC(pDC),ofchronichepatitisBpatientswithhealthyvolunteers. No differences in percentages of mDC and pDC in peripheral blood mononuclear cellswereobservedbetweenchronichepatitisBpatientsandhealthycontrols.The allostimulatorycapacityofisolatedandin vitromaturedmDC,butnotofpDC,was significantlydecreasedinpatientscomparedtocontrols.Accordingly,adecreased percentage of mDC expressingCD80 andCD86 was observed after maturation, comparedtocontrols.Inaddition,mDCofpatientsshowedareducedcapacityto producetumornecrosisfactorαafterastimuluswithsyntheticdoublestrandedRNA andinterferonγ.PurifiedpDCfrompatientsproducedlessinterferonα,animportant antiviral cytokine, in response to stimulation with Staphylococcus aureusCowan strain I than pDC isolated from controls. In conclusion, we show that mDC and pDCarefunctionallyimpairedinpatientswithchronichepatitisB.Thismightbean importantwaybywhichHBVevadesanadequateimmuneresponseleadingtoviral persistenceanddiseasechronicity.

18 IMPAIREDDENDRITICCELLFUNCTION

Introduction

HepatitisBvirus(HBV)infectionrepresentsanenormoushealthproblemworldwide. Todaymorethan350millionpeoplearechronicallyinfectedwithHBVandareatrisk todeveloplivercirrhosisorhepatocellularcarcinoma.Uptillnowitisunclearwhyan individualdevelopsachroniccarrierstate.However,aninadequateimmuneresponse ofthehostisthoughttoplayacriticalroleinthechronicityoftheinfection1.Torecover fromanacuteHBVinfectionbothastronghumoralandcellularimmuneresponse isrequired.Duringanacuteinfectionpatientsexhibitamultispecificandpolyclonal cytotoxicTcell(CTL)responseandastrongtype1Thelpercell(Th1)response2,3. SuchHBVspecificTcellresponsesaregenerallyundetectableinchronicpatients4,5. Dendriticcells(DC)representthemostpotentantigenpresentingcells,andthusplay animportantroleintheinductionofspecificTcellresponses 6.Functionaldefects inDCcouldthereforebeanimportantmechanismofvirustoevadehostimmune responses. In several chronic viral infections, such as human immune deficiency virus1andhepatitisC,impairedfunctionofDChasbeendemonstrated711.ForHBV evidenceexistsaswell,thatshowsimpairedDCfunctioninchronicpatients12,13.Data onthefunctionalityofDCinchronicHBVpatientswasobtainedwithinvitrogenerated monocyte derived DC (moDC).Recently, Osugi et al showed several differences betweenmoDCandtheinvivopresentmyeloidDC 14.Thereforeitwouldbemore accuratetoisolateandstudythefunctionalityofDCprecursorspresentinvivo.Two majorDCprecursorsarethemyeloid(mDC)andplasmacytoid(pDC)dendriticcells15, 16.MDCarecharacterizedbytheabsenceofsurfaceexpressionoflineagemarkers, thepresenceofmarkerssuchasCD1c(blooddendriticcellsantigen1(BDCA1))and CD11c.InresponsetobacterialcompoundsorCD40ligand,mDCcanproducelarge amountsofIL12andtheyrequirethepresenceofgranulocyt/macrophagecolony stimulatingfactor(GMCSF)forsurvival15. PDC,ontheotherhand,arecharacterizedbythenegativityoflineagemarkersand thepresenceofBDCA2,BDCA4andCD123(IL3receptorαchain),theyrequirethe presenceofIL3forsurvivalandcanproducehighamountsoftypeIinterferon(IFN), IFNαandIFNβ,uponexposuretovirusesaswellasbacterialcomponentssuchas CpGoligonucleotides 1719.ThereleaseofIFNαβinitiatesacascadeofeventthat eventuallyleadstotheeliminationofthevirus.Itcanactonseveralcellsandturnon biochemicalpathwaysthatrestrictviralreplicationandrenderhostcellsresistantto furtherviralinfection20.Moreover,typeIIFNisfrequentlyusedastherapyforchronic HBVpatientsandleadstodiseaseremissioninapproximately20%to35%ofcases 21,22.Therefore,theroleoftypeIIFNproducingpDCcouldbeofgreatinterestinthe pathogenesisofchronicHBVinfection. Theaimofthisstudywastodeterminethenumber,phenotypeandfunctionalityof mDCandpDCprecursorsubsetsinperipheralbloodofchronicHBVpatientsandto comparethesecharacteristicswiththoseofhealthyvolunteers.

19 CHAPTER2

Material and Methods

Patients and controls Peripheralheparinizedbloodsampleswereobtainedfrom30patientswithchronic hepatitisB(table1).Allpatientswerenegativeforantibodiesagainsthumanimmune deficiency virus, hepatitisCand hepatitis D. None of the patients was treated for chronicHBVinfectionorreceivedanyothermedication6monthsprevioustoblood sampling.Allpatientshadbiopsyprovenchronichepatitiswithminimaltomoderate fibrosis.The median serumHBVDNA load was 1.1x108 geq/ml (range 1.0x103 –1.2x1010)andmedianalaninetransaminaselevel(ALT)was48U/l(range9–243). FifteenpatientswereHBVenvelopeantigen(HBeAg)positiveand15patientswere HBeAgnegativeandhadantibodiestoHBeAg.Acontrolgroup,matchedforage, genderandrace,comprisedof19healthysubjectswhohadnoevidenceofexposure toHBV(HBVsurfaceantigen(HBsAg)negative).Thestudywasapprovedbythe local ethics committee and all patients and controls in the study gave informed consentbeforeblooddonation.

Table 1. Patient characteristics

All patients (n=30) Controls (n=19)

Sex (male/female) 19 / 11 13 / 6

Age (years)* 39 (19-69) 29 (22-43)

ALT (units/L)* 48 (9-243) n.t.

8 * 1.1x10 HBV-DNA (geq/mL) 3 10 - (1.0x10 -1.2x10 )

HBeAg (+)/anti-HBeAg (+) 15 / 15 - n.t. not tested * median (range)

Virological assessments SerumHBsAg,HBeAg,andantiHBeweredeterminedquantitativelyusingtheIMX system (Abbot Laboratories, NorthChicago, IL) according to the manufacturers instructions. SerumHBVDNA wasdeterminedusingaHBVmonitorassay(Roche AppliedScience,Penzberg,Germany;detectionlimit1x103geq/ml).Whentheserum HBVDNA wasbelow1x103geq/mlHBVDNA theassaywasrepeatedusinganin housedevelopedTaqManPCR(detectionlimit373geq/ml)23.TaqManPCRwasalso

20 IMPAIRED DENDRITIC CELL FUNCTION used for determination of HBV-DNA in the DC subtypes. DNA was extracted from a pellet of 2x104 DC. The pellet was resuspended in 190 µL double distilled water, to which 10 µl of a known amount of internal control, consisting of a seal herpes virus, was added. This material was extracted on a MagnaPure LC isolation system (Roche Applied Science) using the total nucleic acid isolation . Twenty µl of the isolated nucleic acid was used to detect HBV-DNA. Values for the internal control had to be within a range previously determined within the laboratory.

Analysis of myeloid and plasmacytoid dendritic cells subsets in peripheral blood Peripheral blood mononuclear cells (PBMC) were obtained by Ficoll-Isopaque gradient centrifugation. PBMC were incubated for 5 minutes with human immunoglobulins (Octagam, 1.3 mg/ml; Octapharma, Lachen, Switzerland) in PBS containing 1% w/v bovine serum albumin (BSA) before adding specific antibodies to determine the percentage of mDC and pDC. For mDC, PBMC were incubated with a cocktail of FITC-conjugated monoclonal antibodies to the lineage markers CD3, CD14, CD16, CD19, CD20 and CD56 (Becton Dickinson, San Jose, CA), anti-BDCA1 (CD1c)- PE (Miltenyi Biotec, Bergisch Gladbach, Germany) and anti-CD11c-APC (Becton Dickinson). To determine the percentage of pDC, PBMC were incubated with the FITC conjugated lineage marker cocktail, anti-BDCA4-PE and anti-CD123-biotin (Becton Dickinson), and subsequently a secondary step with strepavidin-PerCP (Becton Dickinson). As controls, cells were stained with corresponding isotype-matched control monoclonal antibodies. Stained cells were analyzed using a four-color flow cytometer (FACScalibur, Becton Dickinson) and CellQuest software.

Isolation of myeloid and plasmacytoid dendritic cells MDC (BDCA1+) and pDC (BDCA4+) were isolated from PBMC by positive immunomagnetic selection using the mini-MACS system (Miltenyi Biotec) according to the manufacturers instructions. Briefly, BDCA1+ mDC were isolated by incubating CD19 depleted cells with a PE-conjugated monoclonal antibody (mAb) to BDCA1 followed by anti-PE magnetic beads and separation over a MS-column. In addition, BDCA4+ pDC were isolated by after incubation with a PE-conjugated mAb to BDCA4 followed by anti-PE magnetic beads. Cells were resuspended in culture medium consisting of RPMI 1640 (Bio Whittaker, Verviers, Belgium) containing 10% fetal calf serum (Hyclone, Logan, UT). The isolated mDC and pDC were analyzed for purity by flow cytometry as mentioned in the previous section (mDC; BDCA1+, lineage markers-, CD11c+ and pDC; BDCA4+, lineage markers-, CD123high) and only used if >90% pure.

Expression of cell surface molecules on the surface of myeloid and plasmacytoid dendritic cells by flowcytometry Freshly isolated mDC and pDC (1x104 cells) were incubated with CD80-FITC (Immunotech, Marseille, France), human leucocyte antigen (HLA)-DR-PerCP (Becton Dickinson) and CD86-APC (Becton Dickinson). As a control cells stained with

21 CHAPTER 2 corresponding isotype-matched control monoclonal antibodies were used. Cells were analyzed on the flow cytometer and the mean fluorescence intensity and percent positive stained cells was determined.

Analysis of T cell stimulatory capacity of myeloid and plasmacytoid dendritic cells in a mixed lymphocyte reaction Purified mDC and pDC were matured for 24 hours in 96-well flat bottom culture plates at different concentrations (1.25, 2.5, 5 and 10x103 cells/200 µl) in culture medium containing IL-1β (50 ng/ml; Strathmann Biotech, Hannover, Germany) and TNF-α (25 ng/ml; Strathmann). For mDC GM-CSF (500 U/ml; Leucomax, Novartis Pharma, Arnhem, The Netherlands) and for pDC IL-3 (10 ng/ml; Strathmann) was added as a growth supplement. The next day culture supernatant was removed and nylon-wool purified T cells from a normal healthy volunteer (1.5x105 cells/200 μl) were added to the DC. After 5 days, cell proliferation was assessed by the incorporation of [3H]thymidine (Radiochemical Centre, Amersham, Little Chalfont, UK), 0.5 μCi/well was added and cultures were harvested 18 hours later. Phytohemagglutin 5 μg/ml (Murex, Paris, France) was added to T cells as a positive control.

Cytokine production of stimulated myeloid and plasmacytoid dendritic cells Purified mDC were stimulated at a concentration of 4x104 cells/200 μl in 96 flat bottom Costar plates (Costar, Cambridge, MA) in culture medium with synthetic double-stranded (ds)RNA, polyriboinosinic-polyribocytidylic acid (poly (I:C), 20 μg/ ml; Sigma-Aldrich, St. Louis, MA) in combination with recombinant human IFN-γ (1000 U/ml; Strathmann) and in the presence of GM-CSF (500 U/ml). Purified pDC were stimulated at a concentration of 2x104 cells/200 μl with Staphylococcus aureus Cowan strain I (SAC; 75 μg/ml; Calbiochem, San Diego, CA) and in the presence of IL-3 (10 ng/ml). Cells were cultured at 37°C and 5% CO2 in a humified incubator. Supernatants were harvested after 24 hours. The level of IL-12p70 (Diaclone, Besançon, France) and IFN-α (Biosource International) were determined by standard ELISA according to the manufactures instructions. The levels of TNF-α, IL-6 and IL- 10 were determined by specific solid-phase sandwich ELISA as previously described, using pairs of monoclonal antibodies and recombinant cytokine standards from Biosource International 24, 25.

Statistical analysis Data are expressed as mean ± SEM, unless indicated otherwise. Data were analyzed with SPSS 11.5 for Windows (SPSS, Chicago, IL) using the Mann-Whitney test to compare variables between two independent groups. In all analyzes a p-value of <0.05 was considered statistically significant. Correlations were determined using the Spearman’s correlation test. HBV patients were compared to healthy controls. To analyze the results on the basis of serum viral load and ALT, we categorized all HBV patients according to: 1) HBV-DNA > 108 geq/ml (high, n=16), HBV-DNA 105 to 108 geq/ml (intermediate, n=5), HBV-DNA < 105 geq/ml (low, n=9), and 2) ALT normal

22 IMPAIREDDENDRITICCELLFUNCTION

(upperlimitofnormal(ULN)is35U/L,n=11),ALTnormalto2xULN(intermediate, n=9)andALT>2xULN(high,n=10).

Results

Myeloid and plasmacytoid dendritic cells percentages in peripheral blood are similar in chronic HBV patients and healthy controls TodeterminewhetherachronicHBVinfectionaffectsthefrequenciesofmDCand pDCinperipheralblood,thepercentagesofbothcelltypesinPBMCweredetermined byflowcytometry.MDCwererecognizedinperipheralbloodbystainingwithacocktail oflineagemarkers(CD3,CD14,CD16,CD19,CD20andCD56)andantiBDCA1(Fig 1A).PDCwerecharacterizedbystainingwithantiBDCA4andantiCD123(Fig1B). NosignificantdifferencesinthefrequenciesofbothmDCandpDCwereobserved betweenchronicHBVpatientsandhealthycontrols(Fig1C). 3PerCP 2 LinFitc CD1

A BDCA1PE B BDCA4PE

controls HBV patients Fig.1. PercentagesofmDCandpDC 1 inPBMCweredeterminedwithFACS.

0,9 A)FACSdotblotshowingmDC(gated)

0,8 negative for FITC labeled lineage

0,7 markers and positive for BDCA1PE.

C 0,6 B)FACSdotblotshowingpDC(gated) M PB 0,5 positive for BDCA4PE andCD123 f o

% 0,4 PerCP.C)Nosignificantdifferencesin

0,3 thepercentagesofbothmDCandpDC

0,2 were observed between chronicHBV

0,1 patients (n=30) and healthy controls

0 (n=19). Data are expressed as mean mDC pDC C ±SEM.

23 CHAPTER2

Myeloid dendritic cells of chronic HBV patients are inhibited in their capacity to express costimulatory molecules upon in vitro maturation Freshly isolated DC precursor populations from peripheral blood of patients and controlsexpress onlylowamountsofthecostimulatorymoleculesCD80andCD86. Afterstimulationfor24hoursinthepresenceofIL1βandTNFα,thepercentages of mDC and pDC expressingCD80 andCD86 as well as the mean fluorescence intensitiesofthepositivecellsweresignificantlyincreased.MaturationofmDCwas significantlymoreincreasedcomparedtopDCasisshowninFACShistogramsof onerepresentativehealthycontrol(p<0.001,Fig2).Nodifferenceswereobserved inthemeanfluorescenceintensitiesofCD80andCD86onmDCfrompatientsand controls.However,thepercentagesofmaturemDCexpressingCD80andCD86after stimulation were significantly reduced in the patient group (p<0.05,Table2).The percentagesofpDCexpressingCD80orCD86ofpatientsandcontrolsweresimilar bothbeforeandafterstimulation.Asignificantincreaseinmeanfluorescenceintensity ofCD86onpDCwasfoundintheHBVpatientgroupafterstimulation(p<0.05).The HLADR mean fluorescence intensities were significantly increased on mDC and pDCafterstimulation(p<0.001;Fig2andTable2).NodifferenceinHLADRmean fluorescenceintensitywasfoundwhencomparingpatientstocontrols.

Fig2. FACShistogramsshowingthemeanfluorescenceintensitiesofCD80,CD86 andHLADR of freshly isolated DC (gray histogram) and DC that were matured for24h.withIL1βandTNFα(blackhistogram)andisotypecontrol(dottedline). Resultsfromonerepresentativehealthycontrolareshown.Themeanfluorescence intensities ofCD80,CD86 andHLADR expression were increased after24 h. maturationespeciallyonmDC(A)andtoalesserextentonpDC(B).

24 IMPAIREDDENDRITICCELLFUNCTION

Table 2. Phenotypic characteristics of mDC and pDC

Freshly isolated DC Matured DC DC Markers Patients Controls Patients Controls % 98 ± 2.2 99 ± 0.6 96 ± 12 99 ± 3.1 MDC HLA-DR MFI 1374 ± 78 1476 ± 80 3669 ± 352 4016 ± 479 % 2.6 ± 0.8 1.6 ± 0.6 74 ± 4.1* 84 ± 2.9 CD80 MFI 8.8 ± 1.2 7.2 ± 0.3 64 ± 6.3 66 ± 4.1 % 26 ± 3.9 27 ± 5.6 81 ± 4.8* 92 ± 1.2 CD86 MFI 48 ± 5.1 46 ± 4.1 320 ± 41 311 ± 44 % 95 ± 4.3 97 ± 4.0 96 ± 7.6 98 ± 3.5 PDC HLA-DR MFI 537 ± 39 587 ± 21 1644 ± 164 1579 ± 175 % 1.8 ± 0.5 1.6 ± 0.7 54 ± 3.4 51 ± 4.0 CD80 MFI 6.8 ± 0.5 5.9 ± 0.2 61 ± 5.7 52 ± 3.9 % 2.8 ± 0.9 3.0 ± 1.4 31 ± 5.1 30 ± 7.4 CD86 MFI 41 ± 25 14 ± 1.7 78 ± 9.3* 51 ± 6.2 * p<0.05 versus controls

The allostimulatory capacity of myeloid dendritic cells of chronic HBV patients is impaired ToasseswhethertheTcellstimulatingcapacityofthetwoDCsubsetsisaffectedin chronicHBVpatients,westudiedtheabilityofDCtostimulateTcellsintheallogeneic mixedlymphocytereaction.MaturemDCandpDC(obtainedasdescribedabove) wereaddedatdifferentconcentrationstoTcellsofahealthythirdparty.Fig3Ashows that mDC of patients were less efficient in inducingTcell proliferation than mDC isolatedfromcontrolsatallratiostested(p<0.05).PDCwerecapableofinducingT cellproliferation,butwerelesspotentthanmDC.Thiswasalsoreflectedbytheirlower expressionofHLADRandcostimulatorymolecules,CD80andCD86.Nodifference wasfoundintheTcellstimulatoryfunctionofpDCfrompatientsandcontrols(Fig 3B).BackgroundproliferationofTcellsonlywas<500cpm.TheproliferationofPHA stimulatedTcellswassimilarinbothgroups(datanotshown).

Impaired TNF-α production by myeloid dendritic cells and IFN-α production by plasmacytoid dendritic cells of chronic HBV patients MDC are main producers of IL12 and pDC of IFNα.Chronicity ofHBV infection isthoughttobemainlycausedbyareducedTh1response,possiblycausedbya reduced IL12 and/or IFNα production by DC.To investigate the capacity of the twosubtypesofDCtoproducecytokines,mDCwerestimulatedwithpolyI:Cand IFNγ to induce high IL12p70 production and pDC were stimulated with SAC for highIFNαproduction.Twentyfourhourslaterculturesupernatantswereharvested and analyzed for the production of several cytokines using specificELISAs.The productionofIL12p70,IL10andIL6bymDCwasnotsignificantlydifferentwhen comparingpatientswithcontrols(Fig4AC).However,thesecretionofTNFαinthe supernatantofstimulatedmDCisolatedfromthepatientswassignificantlyreduced compared with healthy controls (Fig 4D). IFNα was not detectable in the culture supernatantofstimulatedmDC(datanotshown). SACstimulatedpDCofpatientsshowedasignificantlyimpairedproductionofIFNα

25 CHAPTER2 comparedtocontrols(p<0.05;Fig5A).IL10productionbypDCwasincreasedinthe patientsalthoughnotsignificantly(p=0.1,Fig5B).Therewasnosignificantdifference inIL6andTNFαproductionofSACstimulatedpDCofcontrolsandpatients(Fig 5CD).OnlylowamountsofIL12p70weredetectableinSACstimulatedpDC(data notshown).

controls HBV patients controls HBV patients

100000 30000

) * m m) p p c

80000 c ( (

n n o o i i t t a a 20000 r r o * o

p 60000 p r r o o c c n n i i

e e n

* n i 40000 i d d i m

mi 10000 y y h h T T *

] ]

H 20000 H 3 3 [ [

0 0 1250 2500 5000 10000 1250 2500 5000 10000 DC/well DC/well A B Fig3. AllostimulatorycapacityofperipheralDCsubtypes.PrecursormDCandpDC were matured for24 h. with IL1β andTNFα. Subsequently DC were cultured at differentnumberswithTcellsfromathirdparty.After5days,thecellswerepulsed foranother18h.with[3H]thymidine.A)mDCofchronicHBVpatients(n=27)showed a significantly reduced capacity to stimulate allogeneicTcells at all ratios tested (*p<0.05) compared to healthy controls (n=15). B) No difference was found in allostimulatorycapacityofpDCofpatients(n=29)andhealthycontrols(n=15).Data areexpressedasmean±SEMcountsperminute(CPM).

40 3000 A B 20 1500

0

l 0

m IL-6 / IL-10 g p 1000 C 20 D 500 10 * 0 0 IL-12 TNF-alpha Fig 4.Cytokine production by isolated peripheral precursor mDC of chronicHBV patients (n=25) and healthy controls (n=14) after stimulation with poly (I:C).After 24h.stimulationcytokineproductionwasdeterminedintheculturesupernatantby specificELISAs.NodifferencewasdetectedintheproductionofA)IL6,B)IL10 andC)IL12p70productionbetweenpatientsandhealthycontrols.D)PurifiedmDC ofpatientsshowedasignificantlyreducedcapacitytoproduceTNFαcomparedto healthycontrols.Dataareexpressedinmean±SEM,*p<0.05.

26 IMPAIREDDENDRITICCELLFUNCTION

1500 100 A B 750 50

0 0 l m

/ IL-6 IL-10 g p

3000 C 6000 D

1500 3000 *

0 0 IFN-alpha TNF-alpha Fig 5.Cytokine production by isolated peripheral precursor pDC of chronicHBV patients (n=23) and healthy controls (n=15) after stimulation with SAC.After24 h stimulationcytokineproductionwasdeterminedintheculturesupernatantbyspecific ELISAs. No difference was detected in the production ofA) IL6, B) IL10 and D) TNFαproductionbetweenpatientsandhealthycontrols.C)PDCofpatientswere significantly impaired to produce IFNα compared to healthy controls. Data are expressedinmean±SEM,*p<0.05.

Dendritic cell function in relation to viral load and alanine transaminase levels WhenanalyzingtheobtainedresultsoftheHBVpatientsinrelationtotheviralloadand ALT,wefoundthatthecapacityofmDCtoproduceTNFαwassignificantlyreduced inpatientswithlowviralloadascomparedtopatientswithhighviralload(p<0.05,Fig 6A).NorelationwasobservedbetweenTNFαproductionandALTlevels. TheIFNαproductionofpDCwassignificantlydecreasedinpatientswithhighALT ascomparedtopatientswithnormalALT(p<0.05,Fig6B).Althoughallpatientswith highALTexhibitedHBVDNAlevelsabove108geq/ml,norelationwasfoundbetween serumviralloadandIFNαproduction. The expression of costimulatory molecules before and after maturation, the allostimulatorycapacityandthecapacitytoproducecytokinesbymDCandpDCwere notrelatedtoviralloadandALT. To exclude that the observed effects are the result of liver inflammation rather than a specific effect of the virus, we studied a separate group of patients with chronicinflammatoryliverdiseaseofnonviralorigin(primarybiliarycirrhosisn=2; hemochromatosis n=4) and aALT comparable to chronicHBV patients (median 40 U/l, range24110 U/l).The mDC isolated from these patients showed similar expression of costimulatory molecules and also displayed similar allostimulatory capacityashealthycontrols.Furthermore,theproductionofIFNαbyplasmacytoid dendritic cells was not reduced in those patients as compared to healthy controls (datanotshown).

HBV-DNA was detectable in DC precursor subpopulations of chronic HBV patients AfunctionalimpairmentoftheDCsubsetscouldbecausedbythepresenceofvirus intheDC.Toinvestigatethis,HBVDNAwasdeterminedintheDCbytaqmanPCR.

27 CHAPTER2

HBVDNAwasdetectedinmDCof15outof30chronicHBVpatients.In12patients HBVDNAwaspresentinpDC.ThelevelofHBVDNAwassignificantlyhigherinpDC thanmDC,1.2x104±2.9x103geq/mlversus6.7x103±2.2x103geq/mlrespectively (mean ± SEM; p<0.01).The presence ofHBVDNA in the mDC and pDC was correlatedwiththeserumHBVDNAvalues(Spearman’scorrelationsr=0.64,p<0.05 andr=0.60,p<0.05respectively).HBVDNA wasnotdetectedinanyofthecontrol DCtested.NocorrelationwasobservedbetweenHBVDNA presenceinmDCand suppressedallostimulatorycapacityanddecreasedCD80andCD86expressionof maturedmDC(datanotshown).

2000 20 l l m / m / g 1000 g 10 p n * *

0 0 low viral load intermediate high viral load normal ALT intermediate high ALT viral load ALT

TNF-alpha IFN-alpha A B Fig6. A) ThecapacityofmDCtoproduceTNFαwassignificantlyreducedinpatients with low viral load (allHBeAg negative) compared to patients with high viral load (94%HBeAgpositive)B)TheIFNαproductionofpDCwasdecreasedsignificantly inpatientswithhighALTascomparedtothepatientswithanormalALT.Dataare expressedinmean±SEM,*p<0.05.

Discussion

DCareprofessionalantigenpresentingcellsthatareextremelypotentininitiating aprimaryimmuneresponse.TheweakorabsentTcellresponsesfoundinchronic HBVpatientscouldbetheresultofadefectintheDCcompartment.Numbersand/or functionalityofDCsubsetsinthebloodmaybeaffectedbythepresenceofthevirus. ThepresentstudyshowedthatpercentagesofmDCandpDCinperipheralbloodof chronicHBVpatientsandcontrolsweresimilar.Ontheotherhand,thefunctionality ofthetwoDCprecursorsubsetswasaffectedinHBVinfectedpatientscomparedto healthycontrols. In vitro matured mDC isolated from chronicHBV patients exhibit an impaired

28 IMPAIREDDENDRITICCELLFUNCTION

allostimulatorycapacitycomparedwithmDCfromhealthycontrols.Itisunlikelythat thisdifferenceiscausedbythedifferenceintheHLAclassIIallelesmismatches,since theallostimulatorycapacitybetweenpDCofpatientsandhealthycontrolswassimilar. ThereducedTcellstimulatorycapacitymaybeduetothedecreasedupregulationof thecostimulatorymolecules,CD80andCD86,afterin vitromaturationofmDCfrom HBVpatients.However,pDCofpatientsshowedanincreasedexpressionofCD86 afterin vitromaturation,whiletherewasnodifferenceinTcellstimulatorycapacityof pDCofpatientsandcontrols.Thissuggeststhatotherfactors,suchascostimulatory or inhibitory molecules, play a role in the decreased allostimulatory capacity. For exampleCD40hasbeenshowntoserveasacostimulusforTcellactivationand alsoCD40ligationisacriticalstepinthefinalmaturationofDCintofullycompetent APC26. Besidestheexpressionofcostimulatorymoleculesorinhibitorymoleculesalsothe capacityofmDCtoproducecertaincytokinesisimportantfortheTcellstimulatory capacity.MDCofHBVpatientswereimpairedtoproduceTNFα.ThecapacityofmDC toproduceTNFαwasdecreasedsignificantlyinpatientswithlowviralload.Sheron etalshowedthatserumTNFαlevelsandthein vitroproductionofTNFαproduction bylipopolysaccharidestimulatedPBMCwaslowerinHBeAgnegativepatientswith lowviralload27.TNFαhasbeenshowntocontributetoallostimulation,itcanactas anautocrinegrowthfactorforDCinducedTcellproliferation28.Furthermore,TNFα playsanessentialroleinthematurationofDC29.Wedemonstratedthatadditionof exogenousTNFαinducedmaturationofDC.Thiswasmanifestedbytheincreased expressionofcostimulatorymoleculesaftermaturationwithILβandTNFα.However, thematurationofmDCwasreducedintheHBVpatientscomparedtohealthycontrols. ThecapacityofmDCtoproduceTNFαwasmostprominentinpatientswithlowviral load,whiletheallostimulatorycapacityofmDCwascomparableforallHBVpatients. Thisindicatesthattheremaybeanadditionalfactoryettobedeterminedthatalso playsaroleinthereducedallostimulatorycapacityofmDCofHBVpatients. WeshowedthatIL12productionbymDCwasnotdifferentbetweenHBVpatients and controls in contrast to previous studies using moDC. In previous studies a reducedallostimulationcorrespondedwithareducedIL12productioninchronicHBV patients12,13.InastudybyLohret al,thereducedTcellresponseinducedbymoDC fromchronicHBVpatientscouldberestoredbyexogenousIL1230.Takentogether theresultsonIL12productionshowthatmDCareclearlydifferentfrommoDC,as alsopreviouslyshownbyOsugiet al 14. In addition to the impaired function of mDC in chronicHBV patients we found a stronglyreducedIFNαproductionbypDCfromchronicHBVpatients.Furthermore, atrendtowardsupregulationofthecytokinesIL6,TNFαandIL10wasobserved in these patients, indicating an imbalance in cytokine production by pDC ofHBV patients.IFNαisapleiotropicmodulatorofhostresistanceandplaysacriticalrolein theinductionofanantiviralstate19,31.Inotherviralinfectionssuchashumanimmune deficiencyvirus1,alsoareducedcapacityofpDCtoproduceIFNαhasbeenshown 32,33.Furthermore,ithasbeenproposedinhumanimmunedeficiencyvirusinfection

29 CHAPTER2 thatpDCcontrolviralreplicationthroughproductionofIFNα34.Thisantiviraleffect of IFNα may also play a role in chronicHBV infection, since we found a more reduced IFNα production in patients with active disease (highALT and high viral load).Recently,IFNαhasalsobeenshowntoplayaroleinplasmacelldifferentiation and subsequent antibody production 35.Therefore, reduced IFNα production may influencetheproductionofHBVspecificantibodies.ExogenousIFNαtherapymay restore the disturbed cytokine environment and could be successful in especially thosepatientswithimpairedendogenousIFNαproduction.Thisissueissubjectof furtherstudies. OurresultsindicatethatbothcirculatingDCsubsetsinperipheralbloodofpatients withchronicHBVinfectionareaffectedintheirfunction.Thisfunctionalimpairmentof DCwasnotobservedinpatientswithchronicinflammatoryliverdiseaseofnonviral origin.Therefore,ourfindingsappearindeedrelatedtoHBVandarenotsolelythe consequenceofinflammationoftheliver.TheDCdysfunctioninchronicHBVpatients maybecauseddirectlybyviralinfectionofDC,interferingwithsignalingpathways involvedinmaturationandcytokineproduction.WeshowedthepresenceofHBVDNA inbothDCsubsets.OthershaveshownthatPMBCandpurifiedcellsubsetscontain HBVDNAandalsoviralintermediates,suggestiveofviralreplication36,37.Moreover, viralparticlesandHBVreplicationintermediateswerefoundinmoDCofchronicHBV patients12,13anddirectinfectionofDChasbeendescribedforotherviruses3840.HBV mayalsointeractwithDCthroughbindingtothecellsurfacereceptors,aspreviously shown for human immunodeficiency virus and hepatitisCinfection 41. From our resultswecannotexcludethatthepresenceofHBVDNAinbothDCsubsetsreflects attachmentofthevirustothecellsurfaceoruptakeofthevirusasabiologicalfunction ofDC.ThevirusmayalsoindirectlyaffecttheDCsubsets,forinstancebyinfectionof othercellswhichinturnproducecytokinescausingachangeincytokineenvironment intheliverorlymphoidorgans.Furtherresearchshoulddeterminethemechanismby whichHBVinterfereswiththeDCfunctionsandwhethertheHBVDNAwefoundin theDCrepresentsreplicatingvirus. ThequestionremainswhetherthechronicHBVinfectioninpatientsisthecauseof apreexistingdysfunctionoftheDCsubtypesorthattheinfectionitselfcausesthe dysfunctionofDC.RecentlyastudyonhepatitisCinchimpanzeessuggestedthat theDCimpairmentisratheraconsequenceofpersistentandactivehepatitisCvirus infectionassociatedwithdiseaseprogression42.Researchstudyingthefunctionality of DC before and after successful therapy forHBV may provide answers for this issue. Inconclusion,ourresultsshowthatbothmDCandpDCarefunctionallyimpairedin chronicHBVpatients.ThiscouldbeoneofthereasonsfortheabsentorweakTcell responsesandthesubsequentcontinuedHBVreplicationinthesepatients.

30 IMPAIREDDENDRITICCELLFUNCTION

References 1. ChisariFV,FerrariC.HepatitisBvirusimmunopathogenesis.Annu.Rev.Immunol.1995;13:2960. 2. Maini MK, BoniC, Ogg GS, et al. Direct ex vivo analysis of hepatitis B virusspecificCD8(+)Tcells associatedwiththecontrolofinfection.Gastroenterology1999;117:138696. 3. WebsterGJ,ReignatS,MainiMK,etal.IncubationphaseofacutehepatitisBinman:dynamicofcellular immunemechanisms.Hepatology2000;32:111724. 4. MarinosG,TorreF,ChokshiS,etal.InductionofThelpercellresponsetohepatitisBcoreantigenin chronic hepatitis B: a major factor in activation of the host immune response to the hepatitis B virus. Hepatology1995;22:10409. 5. MainiMK,BoniC,LeeCK,etal.TheroleofvirusspecificCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.J.Exp.Med.2000;191:12691280. 6. BanchereauJ,SteinmanRM.Dendriticcellsandthecontrolofimmunity.Nature1998;392:24552. 7. DonaghyH,GazzardB,GotchF,PattersonS.Dysfunctionandinfectionoffreshlyisolatedbloodmyeloid andplasmacytoiddendriticcellsinpatientsinfectedwithHIV1.Blood2003;101:450511. 8. AuffermannGretzingerS,KeeffeEB,LevyS.Impaireddendriticcellmaturationinpatientswithchronic,but notresolved,hepatitisCvirusinfection.Blood2001;97:31713176. 9. BainC,FatmiA,ZoulimF,ZarskiJP,TrepoC,InchauspeG.Impairedallostimulatoryfunctionofdendritic cellsinchronichepatitisCinfection.Gastroenterology2001;120:512524. 10. KantoT,HayashiN,TakeharaT,etal.Impairedallostimulatorycapacityofperipheralblooddendriticcells recoveredfromhepatitisCvirusinfectedindividuals.JImmunol1999;162:558491. 11. SteinmanRM,GranelliPipernoA,PopeM,etal.Theinteractionofimmunodeficiencyviruseswithdendritic cells.CurrTopMicrobiolImmunol2003;276:130. 12. BeckebaumS,CicinnatiV,DworackiG,etal.ReductionintheCirculatingpDC1/pDC2RatioandImpaired FunctionofExVivoGeneratedDC1InChronicHepatitisBInfection.Clin.Immunol.2002;104:138. 13. ArimaS,AkbarSM,MichitakaK,etal.Impairedfunctionofantigenpresentingdendriticcellsinpatients withchronichepatitisB:localizationofHBVDNAandHBVRNAinbloodDCbyinsituhybridization.IntJ MolMed2003;11:16974. 14. OsugiY,VuckovicS,HartDN.MyeloidbloodCD11c(+)dendriticcellsandmonocytederiveddendriticcells differintheirabilitytostimulateTlymphocytes.Blood2002;100:285866. 15. LiuYJ,KanzlerH,SoumelisV,GillietM.Dendriticcelllineage,plasticityandcrossregulation.NatImmunol 2001;2:5859. 16. O’Doherty U,Peng M, Gezelter S, et al.Human blood contains two subsets of dendritic cells, one immunologicallymatureandtheotherimmature.Immunology1994;82:48793. 17. BauerM,RedeckeV,EllwartJW,etal.BacterialCpGDNA triggersactivationandmaturationofhuman CD11c,CD123+dendriticcells.JImmunol2001;166:50007. 18. SiegalFP,KadowakiN,ShodellM,etal.Thenatureoftheprincipaltype1interferonproducingcellsin humanblood.Science1999;284:18351837. 19. CellaM,FacchettiF,LanzavecchiaA,ColonnaM.Plasmacytoiddendriticcellsactivatedbyinfluenzavirus andCD40LdriveapotentTH1polarization.NatImmunol2000;1:30510. 20. KatzeMG,HeY,GaleM,Jr.Virusesandinterferon:afightforsupremacy.NatRevImmunol2002;2:675 87. 21. JanssenHL,GerkenG,CarrenoV,etal.InterferonalfaforchronichepatitisBinfection:increasedefficacy ofprolongedtreatment.TheEuropeanConcertedActiononViralHepatitis(EUROHEP).Hepatology1999; 30:23843. 22. SchalmSW,HeathcoteJ,CianciaraJ,etal.Lamivudineandalphainterferoncombinationtreatmentof patientswithchronichepatitisBinfection:arandomisedtrial.Gut2000;46:562568. 23. Pas SD, FriesE, De ManRA, OsterhausAD, NiestersHG. Development of a quantitative realtime detectionassayforhepatitisBvirusDNA andcomparisonwithtwocommercialassays.JClinMicrobiol 2000;38:2897901. 24. KalinskiP,SchuitemakerJH,HilkensCM,KapsenbergML.ProstaglandinE2inducesthefinalmaturation ofIL12deficientCD1a+CD83+dendriticcells:thelevelsofIL12aredeterminedduringthefinaldendritic cellmaturationandareresistanttofurthermodulation.JImmunol1998;161:28049. 25. SnijdersA,HilkensCM, van derPouw KraanTC,Engel M,Aarden LA, Kapsenberg ML.Regulation of bioactive IL12 production in lipopolysaccharidestimulated human monocytes is determined by the expressionofthep35subunit.JImmunol1996;156:120712. 26. McLellanAD,SorgRV,WilliamsLA,HartDN.HumandendriticcellsactivateTlymphocytesviaaCD40: CD40liganddependentpathway.EurJImmunol1996;26:120410. 27. SheronN,LauJ,DanielsH,etal.Increasedproductionoftumournecrosisfactoralphainchronichepatitis

31 CHAPTER2

Bvirusinfection.JHepatol1991;12:2415. 28. McKenzie JL,Calder VL, Starling GC,Hart DN.Role of tumour necrosis factoralpha in dendritic cell mediatedprimarymixedleucocytereactions.BoneMarrowTransplant1995;15:16371. 29. SteinmanRM.Thedendriticcellsystemanditsroleinimmunogenicity.AnnuRevImmunol1991;9:271 96. 30. LohrHF,PingelS,BocherWO,etal.ReducedvirusspecificThelpercellinductionbyautologousdendritic cellsinpatientswithchronichepatitisBrestorationbyexogenousinterleukin12.ClinExpImmunol2002; 130:10714. 31. KadowakiN,AntonenkoS,LauJY,LiuYJ.Naturalinterferonalpha/betaproducingcellslinkinnateand adaptiveimmunity.JExpMed2000;192:21926. 32. Feldman S, Stein D,Amrute S, et al. Decreased interferonalpha production inHIVinfected patients correlates with numerical and functional deficiencies in circulating type2dendritic cell precursors.Clin Immunol2001;101:20110. 33. ChehimiJ,CampbellDE,AzzoniL,etal.Persistentdecreasesinbloodplasmacytoiddendriticcellnumber andfunctiondespiteeffectivehighlyactiveantiretroviraltherapyandincreasedbloodmyeloiddendriticcells inHIVinfectedindividuals.JImmunol2002;168:4796801. 34. SoumelisV,ScottI,GheyasF,etal.Depletionofcirculatingnaturaltype1interferonproducingcellsinHIV infectedAIDSpatients.Blood2001;98:90612. 35. JegoG,PaluckaAK,BlanckJP,ChalouniC,PascualV,BanchereauJ.Plasmacytoiddendriticcellsinduce plasmacelldifferentiationthroughtypeIinterferonandinterleukin6.Immunity2003;19:22534. 36. YoffeB,NoonanCA,MelnickJL,HollingerFB.HepatitisBvirusDNAinmononuclearcellsandanalysisof cellsubsetsforthepresenceofreplicativeintermediatesofviralDNA.JInfectDis1986;153:471477. 37. GuJR,ChenYC,JiangHQ,etal.StateofhepatitisBvirusDNAinleucocytesofhepatitisBpatients.JMed Virol1985;17:7381. 38. BlauveltA,AsadaH,SavilleMW,etal.ProductiveinfectionofdendriticcellsbyHIV1andtheirabilityto capturevirusaremediatedthroughseparatepathways.JClinInvest1997;100:204353. 39. FugierVivier I, ServetDelpratC,RivaillerP,Rissoan MC, LiuYJ,RabourdinCombeC. Measles virus suppressescellmediatedimmunitybyinterferingwiththesurvivalandfunctionsofdendriticandTcells.J ExpMed1997;186:81323. 40. Riegler S,HebartH,EinseleH, BrossartP, Jahn G, SinzgerC. Monocytederived dendritic cells are permissivetothecompletereplicativecycleofhumancytomegalovirus.JGenVirol2000;81:3939. 41. GeijtenbeekTB,vanKooykY.DCSIGN:anovelHIVreceptoronDCsthatmediatesHIV1transmission. CurrTopMicrobiolImmunol2003;276:3154. 42. RollierC,DrexhageJA,VerstrepenBE,etal.ChronichepatitisCvirusinfectionestablishedandmaintained inchimpanzeesindependentofdendriticcellimpairment.Hepatology2003;38:8518.

32 CHAPTER 3

Flow cytometry of fi ne-needle aspiration biopsies: a new method to monitor intrahepatic immunological environment in chronic viral hepatitis

Modifiedfrom:

Flowcytometryoffineneedleaspirationbiopsies:anewmethodtomonitor intrahepaticimmunologicalenvironmentinchronicviralhepatitis D.Sprengers1,R.G.vanderMolen 1,J.G.Kusters1,J.Kwekkeboom1,L.J.W.vander Laan1,H.M.G.Niesters2,E.J.Kuipers1,R.A.DeMan1,S.W.Schalm1,H.L.A.Janssen1. J.ViralHepat2005;12:507512

Coarsevs.FineNeedleaspirationbiopsy D.Sprengers1,R.G.vanderMolen1,J.G.Kusters1,H.L.A.Janssen1 JHepatol2004;41:503504

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands CHAPTER3

Abstract

Informationaboutcharacterandgradeoftheintrahepaticimmuneresponsein viralhepatitisisimportantforevaluationofdiseasestageandef fectoftherapy . Complicationslikehaemorrhageprovidealimitationtofrequentlyperformingtissue needlebiopsies(TB),andhavemademanyuseperipheralbloodassurrogate markerinstead. Thefineneedleaspirationbiopsy(FNAB)oftheliverrepresents asafeandatraumaticmethodthatallowsfrequentcytologicalsampling.Ouraim wastoinvestigatewhetherflowcytometryofFNABspecimensallowscoanalysisof phenotype,functionandspecificityofkeypopulationsofliverinfiltratinglymphocytes (LIL).In20consecutivepatientswithchronicviralhepatitis(10HBV ,10HCV)flow cytometrywasperformedonFNABcytology ,simultaneouslyobtainedlymphocytes isolatedfromaTBandperipheralbloodmononuclearcells(PBMC).TheratioCD8+/ CD4+lymphocytesinFNABcorrelatedwellwithLIL fromTB(r=0.78,p<0.05)but differedfromPBMC(meanratio:2.6;2.1;0.7respectively).Similarly,acorrelationwas observedforpercentageCD56+NKcells(mean%:29.9;32.3;14.5respectively;r= 0.69,p<0.05).ThepercentageIFNγproducingCD3+lymphocytesinbothFNABand TBwashigherthaninPBMC(mean%:41;44;22respectively;p<0.05).Furthermore, tetramericcomplexesallowedanalysisofHBV specificTcellsinFNABspecimens. Inconclusion,flowcytometryofFNABallowseasy,atraumaticandreliableanalysisof lymphocytesobtainedfromtheintrahepaticcompartment. Therefore,theFNABisa valuabletoolinthestudyofimmunopathologyofviralhepatitis,anditmaycontribute toimprovedclinicalevaluationofchronicviralliverdisease.

34 CHAPTER3FLOWCYTOMETRYOFFNAB

Introduction

Manyliverdisorders,suchashepatitisBandCvirusinfection,arecharacterized bychronicinfl ammationinducedbyahostimmuneresponse.Informationabout characterandgradeoftheintrahepaticimmuneresponseisimportantforevaluation ofdiseasestageandeffectoftherapy.Themostwidelyusedliversamplingmethod, andtheacceptedstandard,isthe14Gaugecoretissueneedlebiopsy .Evenwith currentbiopsytechnologyseriouscomplicationslikepain,haemorrhageandbile leakageprovidealimitation,especiallyforrepeateduse.Themortalityrateafter TB isapproximately1in10,000patients 1.Therefore,mosthumanstudiesinvestigating viralhepatitishavefocussedonimmuneef fectorcellsderivedfromperipheral blood(PB).Althoughthisprovidedvaluableinformation,circulatingandintrahepatic hepatitisBvirus(HBV)specificTcellsdisplayeddifferentphenotypeswithregardto theiractivationstatusandthusmaydif ferinef fectorfunctionssuchascytotoxicity andinterferongamma(IFN γ)production 2.Moreover,itwasshownthatresident intrahepaticpopulationsoflymphocytesdif fersignificantlyfrompopulationsinthe circulation,suggestingauniqueimmunologicalenvironmentintheliver3. Thisemphasisestheneedforasafetechniquetoobtainsufficientexvivointrahepatic immuneeffectorcellstomonitortheimmuneresponsedirectedagainstviralhepatitis. Fineneedleaspirationbiopsy(FNAB)hasprovedtobeaneasy ,atraumaticand reliablemethodforthediagnosisofacutere jectioninkidneygraftedpatients 4and morerecentlyinlivertransplantrecipientsandpatientswithfocalliverlesions 57.Up tonowflowcytometryonliverFNABshasconcentratedonevaluating DNAcontent andploidyinthecontextofintraabdominalmasses 8,9.However,FNABmayalsobe avaluabletoolinimmunopathologicalresearchandclinicalpracticeinchronicviral hepatitis.C D56+NaturalKillercells(N Kcells),CD8+cytotoxicTcells(CTL)and CD4+helperTcells(Th)haveallbeenshowntobeessentialintheimmuneresponse againstviralhepatitis 2,10,1 1.The aimofthisstudywastoassesswhetherfl ow cytometricanalysisofphenotypeandfunctionofthesekeypopulationsofimmune effectorcellsobtainedbyFNABandTB,allowequallygoodanalysisofliverinfiltrating lymphocytesinpatientswithchronicHBVandHCVinfection.Todeterminepotential “contamination”oftheFNABaspiratewithperipheralblood,lymphocytesfromFNAB cytology,TBandPBareanalysedsimultaneously.

Methods

Fine-Needle Aspiration Biopsy. Twentyconsecutivepatients(10HBV,10HCV),undergoingTBforchronicHBVorHCV infection,werestudied.TBwasperformedaspartofthediagnosticevaluationand allpatientssignedinformedconsenttoperformaFNAB justbeforetheconventional tissuebiopsywastaken. DetailsoftheFNABprocedurearedescribedelsewhere 12.Briefly,amandarincontaining 25gauge(diameter0.5mm)needle(Braun AG, Melsungen,Germany)ispuncturedinthe 8thor 9thrightintercostalspace. After

35 CHAPTER 3 removal of the mandarin a 10 mL syringe filled with 2 mL RPMI supplemented with 0.1% human serum albumin (Sanquin CLB, Amsterdam, the Netherlands) and 25 IU/mL heparin (Leo Pharma, Breda, the Netherlands) is attached. Liver cells are aspirated by negative syringe pressure. Next, the needle is flushed with 3 mL culture medium to obtain the liver cells from inside the needle and the specimen is transferred into a 15 mL tube and placed on ice immediately. Per patient two FNABs are performed and the aspirates are pooled before further analysis. If at macroscopic evaluation an aspirate contained predominantly blood and only few tissue fragments, an additional specimen was taken immediately. Only representative FNAB-specimens 12, i.e. containing 7 hepatocytes/100 leukocytes in a May-Grünwald-Giemsa stain of a cytospin preparation made from a fraction of the aspirate, were analysed with flow cytometry.

Immunophenotyping and intracellular cytokine staining PBMC were isolated from heparinized blood samples by Ficoll-Hypaque density gradient centrifugation. Erythrocytes were removed from FNAB-specimens by

NH4CL. Liver infiltrating lympocytes (LIL) from TB were isolated after digestion with collagenase type IV (0,5 mg/ml; Sigma Chemical Co., St. Louise, USA (Sigma)) and DNAse (25μg/ml; Gibco, Paisley Scotland) for 30 minutes at 37 oC. The cell suspension was washed twice before antibody staining and four-color flow cytometric evaluation using FACS Caliber (Becton and Dickinson Biosciences; San Jose, USA (BD)) and CELLQuest software (BD). Immunofluorescence staining was performed as described previously 2. To determine IFN-γ production cells were stimulated with phorbol myristate acetate (PMA;50ng/ml; Sigma) and ionomycin (1μM; Sigma) in the presence of Brefeldin (10μg/ml; Sigma) or with Brefeldin only (negative control). The following antibodies were divided in 2 panels for FACS analysis: anti-CD4 PerCP Cy5, -hIFNγ FITC, -CD8 PerCP (BD); anti-CD8 APC (Dako, Glostrup, Denmark (Dako)) anti-CD56 PE, -CD56 APC and –CD3 PE (Immunotech, Marseille, France). Matched isotype antibodies were used for control staining. In a separate experiment it was determined that LIL obtained by FNAB display a similar degree of autofluorescence as lymphocytes from PBMC. Since FNABs contain limited cell numbers the isotype controls were performed with PBMC. A minimum of 30.000 lymphocytes isolated from PB and liver tissue was analysed for each labelling together with all lymphocytes collected with the FNAB.

HLA typing, HLA-A2 tetramer staining Screening for HLA-A2 haplotype positive HBV patients was performed by staining PBMC with an anti-HLA-A2 antibody derived from a hybridoma (clone BB7.2, ATCC HB-82). Staining was followed by an FITC-conjugated rabbit anti-mouse anti-IgG (Dako) secondary antibody and flow cytometric analysis. In HLA-A2+ HBV patients HBV-specific T cells were stained with the soluble HLA-A2 peptide tetramer: HBc 18- 27 (FLPSDFFPSV) APC-conjugated (ProImmune, Oxford, UK) together with anti-CD8 FITC (Dako). Staining was performed as reported before 2. In control experiments with

36 CHAPTER3FLOWCYTOMETRYOFFNAB

4HLAA2positivehealthycontrolsandHLAA2negativeHBVpatientsthetetramer boundto0.019±0.002and0.013±0.004%oftotalCD8+ Tc ellsrespectively. Thereforebackgroundstaininglevelwasdetermined0.025%(mean+2SD).

Statistical analysis ThepairedPearsoncorrelationtestwasusedtocorrelateresultsobtainedbyFNAB toLILi solatedfromTBandlymphocytesfromPB.TheagreementbetweenFNAB andTBwasdeterminedasdescribedbyBlandand Altman13andreportedasB& A pvalues.

Results

FNAB evaluation IncytospinevaluationtheFNABaspiratesof20consecutivepatientswithviral hepatitiswerescoredasrepresentative,i.e.contained7hepatocytes/100leukocytes andwerethusunlikelytobe“contaminated” withlargeamountsofPBMC. ConsequentlyallFNABspecimensweresuitableforanalysisbyflowcytometry.The FNABsamplecontainedamedianof10,042(2,62040,346)lymphocytesthatwere analysedbyflowcytometry.Usingtrypaneblueexclusioncriteriatheviabilitywas >95%.Allsampleswerecollectedwithoutanycomplicationstothepatients.

Ratio CD8 / CD4 CytotoxicTcells(CTL)andhelperTcellshavebothbeenshowntobeessentialinthe immuneresponsetoviralhepatitis 2,14.LymphocytesisolatedfromPBandLI Lfrom FNABandTBofall20patientswereevaluatedforexpressionofCD8, amarkerfor CTL,andforexpressionofCD4,amarkerforhelperTcells.Fig1showsthatLI Lin bothFNABandTBcontainedsigni ficantlymoreCD8+thanCD4+lymphocytesas opposedtoPB.Therewasasigni ficantcorrelationbetweenthetwoliversampling methods.The meanratioCD8+/ CD4+lymphocyteswas0.7(±0.1)inPB, 2.6(± 0.34)inLILfromTBand2.1(±0.3)intheFNABaspirate(FNABvs.TB:r=0.78,p< 0.0001;B&Ap=0.38)(table1).

Table 1. Comparison of key populations of lymphocytes present in FNAB, tissue biopsy and peripheral blood

FNAB Tissue Biopsy PB TB vs. FNAB Ratio CD8+/CD4+ 2.1 [± 0.3] 2.6 [± 0.34] 0.7 [± 0.1] r = 0.78, p < 0.0001 lymphocytes B&A p = 0.38 % CD56+ 32.3% [± 2.9] 29.9% [± 3.3] 14.5% [± 2.4] r = 0.69, p = 0.0002 lymphocytes B&A p = 0.44 % IFNγ+ CD3+ 42% [± 4.6] 39% [± 6.1] 21% [± 3.5] r = 0.79, p = 0.001 lymphocytes B&A p = 0.13

Results represent mean [± SEM] of ratio CD8+/CD4+ lymphocytes, percentage CD56+ lymphocytes and percentage IFNγ+ CD3+ lymphocytes in FNAB-cytology, LIL isolated from tissue biopsy (TB) and lymphocytes from peripheral blood of 20 patients with viral hepatitis.

37 CHAPTER3

A 4 D C r = 0.78 / r = 0.82 8 4 D D C

C / o i 8 t a D r C

d o o i t o l a b r l y a s r p e o h i p i B r e P

FNAB ratio CD8/CD4 FNAB ratio CD8/CD4 1 2

B

FNAB Tissue Biopsy Peripheral Blood

43.7%CD8/CD4: 3.3 60.8% CD8/CD4: 3.427.7% CD8/CD4: 1.1

C

P

A

8

D C

13.2% 17.8% 25.2%

CD4 PerCP-Cy5 Fig1. A) RatioCD8+/CD4+lymphocytesintissuebiopsyversusFNAB(1)and peripheralbloodversusFNAB(2).ResultsofPearsoncorrelationtest. Withthemethodforassessingagreementbetweentwomethodsofclinical measurement,asdescribedbyBlandand Altman,wefindanonsignificant p(p=0.38)in Aa ndasignificantp(p<0.0001)inB. Thisindicatesanon significant(A)andsignifi cant(B)dif ferencebetweenthetwosampling techniques. B) FACSdotblotsofCD8+/CD4+lymphocytesinFNAB,tissuebiopsyand peripheralblood. Thenumberintheupperleftquadrantindicatesthe percentageCD8+lymphocytes. Thenumberinthebottomrightquadrant indicatesthepercentageCD4+lymphocytes. TheratioCD8+/CD4+ lymphocytesisdepictedintheupperrightquadrant.Datashownofa representativepatient(patient8).

Fraction CD56+ lymphocytes AlargeproportionofintrahepaticlymphocytesconsistsofCD3CD56+NaturalKiller cells(N Kcells) 15.Consideringthepotentialimportanceofthesecellsinhepatic immunity,thefractionofCD 56+lymphocyteswasdeterminedinFNABspecimens andTBs.ResultsshowasignificantcorrelationinpercentageCD56+cellsinLILfrom

38 CHAPTER3FLOWCYTOMETRYOFFNAB

FNABsand TBs.The meanpercentageofCD56+lymphocyteswas14.5(±2.4%) inPB,29.9(±3.3%)inLIL isolatedfromtheTBsand32.3(±2.9%)intheFNAB specimens(FNABvs.TB:r=0.69,p=0.006;B&Ap=0.44)(Table1).

IFN-γ production RecentstudieshaveshownthatIFNγsecretionbyTcellsplaysanimportantrolein theantiviralimmuneresponsetoHBV andHCV10.In17patients(20minus3HLA A2+HBVpatients)thepercentageIFN γproducingCD3+ Tcellswasdetermined after5hourstimulationwithPMA andionomycine.InPBameanof21%oftotal CD3+TcellsproducedIFN γversus39%ofthe TcellsisolatedfromtheTBsand 42%ofTcellsintheFNABspecimens(FNABvs.TB:r=0.79,p=0.001;B&A p= 0.13)(Table1).Fig2showsdataofarepresentativepatient.Nosignificantdifference wasfoundbetweenthenumberofIFNγproducingNKcellsinPB,TBandFNAB;the meanswere50%,54%and63%,respectively(FNABvs.TB:r=0.62,p=0.02;B&A p=0.88).

FNAB Tissue Biopsy Peripheral Blood

73%78% 50% e P - 3 D C

IFNg-Fitc Fig2. IFNγproductionbyCD3+lymphocytes. Then umberinthe upperright quadrant indicatesthepercentageIFN γproducingCD3+lymphocytesoftotalCD3+ lymphocytes.Datashownofarepresentativepatient(patient2).Inallcases backgroundIFNγproductionwasnegligible.

HBc 18-27-tetramer binding of CD8+ lymphocytes To investigatewhethertheFNABprovidess ufficientmaterialtoade quatelyanalyse cellsthatarepresentintheliverinverylowfre quencies,inHLAA2+HBVpatients virusspecificTcellswerestainedwithtetramericcomplexesabletovisualiseCD8+T cellstargetedattheHBVcoreepitope1827,2.Threeoutof10chronicHBVpatients wereHLAA2+.In1of3patientstetramerstainingexceededthebackgroundlevelof 0.025%oftotalCD8+ Tcellpopulation.InPBofthispatient0.19%oftotalCD8+ T cellsshowedtetramerbinding,whereasthiswas1.5%inLILisolatedfromtheTBand 1.3%intheFNABaspirate(fig3).

39 CHAPTER3

Multiple sampling SincematerialofmultipleFNABsispooled,samplingerrormayprovidealimitation. Inanadditionalexperimentincluding5patientswithdiffuseliverdisease,we comparedthepercentagesCD4+,CD8+andCD56+lymphocytespresentintwo FNABsthatwereperformedatthesametimepointbutanalysedseparately .The medianmeasurementerrorwas1.2%forCD4,2.2%forCD8and5%forCD56,with highcorrelationinapairedPearsontest(r>/=0.89,p

1,6 1,5

1,4 1,3 s l 1,2 cel

T

+ 1 8 D C / 0,8 + er 0,6 am r et T

0,4 % 0,19 0,2

0 FNAB TB PB Fig3. In3HLAA2+HBVpatientsHBVspecificTcellswerestainedwiththesolubleHLA A2peptidetetramerHBc1827.Inpatient1tetramerpositiveCD8+ Tc ellswere detectableinFNAB,TBandPB.HBc1827tetramerbindinginmaterialfrompatient 4and6didnotexceedbackgroundlevel(datanotshown).

Discussion

Thisisthefi rstreporton flowcytometricimmunophenotypingoftheintrahepatic compartmentusingFineNeedleAspirationBiopsies(FNAB)oftheliver.Weshowthat theFNABaspiratecontainssufficientmaterialforflowcytometryimmunophenotyping experimentsandfunctionalassays ;evenwhenthecellpopulationofinterestis specificandpresentinsmallnumbers,likeHBVspecificCD8+Tcells. ForclinicalpracticeinhepatologytheadvantagesoftheliverFNABarebecoming increasinglyapparent.TheFNABisaneasyprocedurethatismuchbettertolerated thanastandardlivertissuebiopsy.LargeseriesinwhichseveralthousandFNABs wereevaluateddescribeanexcellentsafetyprofi le 12. Uptonowinourclinic we havecollected>500FNABsamplesoftheliverwithoutanyseriouscomplications tothepatient.InvasivenessiscomparabletoavenapunctionandtheFNABcanbe performedatanypatientvisitwithoutanaesthesiaorotherpreparations. Therefore theFNAB,incontrasttoa TB,issuitableforrepetitiveuseasbedsideprocedureor

40 CHAPTER3FLOWCYTOMETRYOFFNAB

intheoutpatientclinic,enablingfrequentmonitoringandimprovedclinicalanalysis oftheintrahepaticimmuneresponse,forinstancetoevaluatetheimpactofantiviral therapyonhepaticinflammatoryinfiltration. InthisstudylymphocytepopulationspresentinFNABspecimenswerecompared tolymphocytesisolatedfromTBandPB.Theoretically,representativenessofFNAB samplescanbeaffectedby“contamination”withPB,inwhichcasetheobtained resultswouldcorrelatewithPBinsteadoftheTBs.Visualinspectionforblood contaminationduringtheFNABprocedureaswellascytospinevaluationofthe FNABaspirateareimportantinthisrespect.RepresentativeFNABspecimens,i.e. containing7hepatocytes/100leukocytesinaMayGrünwaldGiemsastainofafraction oftheaspirate,areanalysedwithfl owcytometry.Thisprocedureisbasedupona comparisonbetweentheinflammatorycellsofliverandbloodsamplesasdescribed forlargenumbersofliverFNABsinthesettingofviralhepatitisandtransplantation 1618.Weshowthatinexperiencedhandsthesecriteriaarealmostnevermet,since innoneofthe20consecutivepatientstheFNABsamplehadtobeexcludedfrom furtheranalysis.AlthoughaMayGrünwaldGiemsastainoftheaspirateisalways analysedinparalleltopreventanalysisofnonrepresentativeFNABs,itcannot fullyexcludemixturewithleukocytesfromPB.Previousstudieshavereportedthat intrahepaticlymphocytepopulationscontainmoreCD8+thanCD4+lymphocytes asopposedtoPB 3,15.Therefore, theratioCD8+/CD4+lymphocytescanbeused asasensitivemeanstovalidatefl owcytometryofFNABinevaluatingintrahepatic immuneresponse.InthisstudytheratioCD8/CD4inFNABscorrelatedwellwiththat observedinTBs,andwassignificantlyhigherthanthatobservedinPB. Asflowcytometricanalysisdoesnotrequirefi xationoflivingcells,FNABcytology maybeusedtoinvestigatethepossibleroleofcytokinesandvirusspecifi cTcells inthepathogenesisofchronicviralhepatitis. To illustratethisapplicationofFNAB weshowthatthepercentagesintrahepaticCD3+lymphocytesproducingIFNγin FNABandTBcorrelatewell.InordertoinvestigatewhetherFNABprovidessufficient materialtoanalysethevirusspecificCD8+Tcellresponse,weusedsolubleHLAA2 tetramericcomplexesabletovisualiseTcellsspecificfortheHBVcoreepitope1827. ComparedtoPB,thereissequesteringoftetramer+CD8+TcellsinLILisolatedfrom TBaswellasinFNABcytologyofapatientwithchronicHBV ,confirmingprevious reports 2.Itthereforeappearsthatamedianof10,000lymphocytesintheFNAB aspirateallowsanalysisofHBVspecificCTL. NotonlydoestheFNABprocedureallowforfl owcytometricanalysisofinfrequent cellpopulations.InthisgenomiceraothertechnologieslikePCRandmicroarray allowreproducibleanalysisofextremelylowcellnumbersthatcaneasilybeobtained byFNAB.Therefore,diagnosticproceduresliketheFNAB,mayalsobeincreasingly importantforunderstandingpotentialintracellularinteractionsbetweenthehostcell andthevirus,thatcontributetopersistenceofviralhepatitis. AlthoughresultsofTBandFNABcorrelatewell,samplingerrormayprovidea limitationinflowcytometricanalysisofFNABssincematerialofmultiplebiopsies ispooled.However,weshowedthatinthisprotocolthesamplingerrorinvolvedin

41 CHAPTER3 evaluationofFNABcytologyisminimal.Infact,theFNABprocedurecouldbeused toperformmultiplepassesindifferentareasoftheliver,thusreducingthesampling errorthatisoneofthelimitationsofthestandardtissueneedlebiopsies. Inconclusion,fl owcytometryoffi neneedleaspirationbiopsyoftheliverallows easy,atraumaticandreliableanalysisoflymphocytesobtainedfromtheintrahepatic compartment.Therefore,theFNABisavaluabletoolinthestudyofimmunopathology ofviralhepatitisthatmaycontributetoimprovedclinicalevaluationofchronicviral liverdisease.

Acknowledgements

Theauthorswishtothan kABertolettifromtheInstituteofHepatology , University CollegeLondon,forhisvaluablehelpanddiscussion.WewouldalsoliketothankT.J. TangforadviceregardingtheFNABprocedureandB.Hansenforherassistancewith thestatisticalanalysis.

42 CHAPTER3FLOWCYTOMETRYOFFNAB

References 1. BravoAA,ShethSG,ChopraS.Liverbiopsy.NEnglJMed2001;344:495500. 2. MainiMK,BoniC,LeeCK,etal. TheroleofvirusspecificCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 3. NorrisS,CollinsC,DohertyDG,etal.Residenthumanhepaticlymphocytesarephenotypicallydif ferent fromcirculatinglymphocytes.JHepatol1998;28:8490. 4. vonWillebrandE,HayryP.Reproducibilityofthefi neneedleaspirationbiopsy .Analysis of93double biopsies.Transplantation1984;38:3146. 5. SchlittHJ,NashanB,RingeB,etal.Dif ferentiationoflivergraftdysfunctionbytransplantaspiration cytology.Transplantation1991;51:78693. 6. FrancaAV, ValerioHM, TrevisanM,etal.Fineneedleaspirationbiopsyforimprovingthediagnostic accuracyofcutneedlebiopsyoffocalliverlesions.ActaCytol2003;47:3326. 7. GreeneCL,FehrmanI, TilleryGW,HusbergBS,KlintmalmGB. Ac leardistinctionbetween“immune activationofrejection”and“noimmuneactivation”inlivertransplantaspirationcytology .TransplantProc 1988;20:6612. 8. FuhrJE,KattineAA,NelsonHS,Jr .Flowcytometryofliverfineneedleaspirates.JSurgOncol1994;56: 1538. 9. GranadosR,GarciaVelaJA,FenandezSegovianoP,etal.FlowcytometricDNA analysisonfineneedle aspirationbiopsiesofliverlesions.Cytopathology2002;13:27383. 10. FreseM,SchwarzleV,BarthK,etal.Interferongammainhibitsreplicationofsubgenomicandgenomic hepatitisCvirusRNAs.Hepatology2002;35:694703. 11. LauGK,SuriD,LiangR,etal.ResolutionofchronichepatitisBandantiHBsseroconversioninhumansby adoptivetransferofimmunitytohepatitisBcoreantigen.Gastroenterology2002;122:61424. 12. LautenschlagerI,HockerstedtK,HayryP.Fineneedleaspirationbiopsyinthemonitoringofliverallografts. TransplInt1991;4:5461. 13. BlandJM,AltmanDG.Statisticalmethodsforassessingagreementbetweentwomethodsofclinical measurement.Lancet1986;1:30710. 14. DayCL,SethNP,LucasM,etal.ExvivoanalysisofhumanmemoryCD4 TcellsspecificforhepatitisC virususingMHCclassIItetramers.JClinInvest2003;112:83142. 15. HataK,V anThiel DH,HerbermanRB,Whiteside TL.Phenotypicandfunctionalcharacteristicsof lymphocytesisolatedfromliverbiopsyspecimensfrompatientswithactiveliverdisease.Hepatology1992; 15:81623. 16. HayryP,vonWillebrandE.Practicalguidelinesforfineneedleaspirationbiopsyofhumanrenalallografts. AnnClinRes1981;13:288306. 17. FranceschiniN,GoncalvesLF,PromptCA,BarrosSG,CerskiCT,CostaCA.Fineneedleaspirativebiopsy oftheliverinHBsAGpositivepatientswithendstagerenalfailure.RenFail1994;16:4919. 18. KuijfML,KwekkeboomJ,KuijpersMA,etal.Granzymeexpressioninfi neneedleaspiratesfromliver allograftsisincreasedduringacuterejection.LiverTranspl2002;8:9526.

43

CHAPTER 4

Different composition of intrahepatic lymphocytes in the immune-tolerance and immune-clearance phase of chronic hepatitis B

D.Sprengers1,R.G.vanderMolen1,J.G.Kusters1,B.Hansen1,H.G.M.Niesters2, S.W.Schalm1,H.L.A.Janssen1.

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

JMedVirol2006;78:561568 CHAPTER4

Abstract

BasedonvirologicalandbiochemicalparameterschronicHBVpatientsare dividedintodistinctclinicalphases:theimmunetolerancephase,theimmune clearancephaseandtheinactivecarrierstate.Uncleariswhetherthesephases havecharacteristicintrahepaticimmuneresponses.W eaimedtocharacterisethe compositionofliverderivedlymphocytesinpatientswithchronicHBVinfection.In47 patientsthecompositionofliverderivedlymphocyteswasanalysedbyflowcytometry ofFineNeedleAspirationBiopsiesoftheliver .The proportionNKcellsintheliver wassignificantlyhigherinimmunetolerantthaninimmuneclearancepatientsand inactivecarriers.NodifferenceswerefoundinproportionCD4+ TcellsandCD8+ Tcells,inthesephases.However,whenpatientsintheimmuneclearancephase, withsimilarALT,weregroupedaccordingtoviralload,theproportionCD8+ Tcells washigherinthosewithhighviralload.Incontrast,theproportionCD4+Tcellswas increasedinpatientswithlowHBVDNA.Thesedifferenceswereabsentinperipheral blood.IntrahepaticHBV specificCD8+ Tcellsweremainlyfoundinimmune clearancepatientswithlowviralload. Inconclusion,cleardifferencesintheintrahepaticcellularinfi ltratewerefound betweenthevariousclinicalphasesofchronicHBVinfection. Thesefindingsare relevanttothedesignofnew,individualisedantiviralstrategies.

46 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

Introduction

HepatitisBvirus(HBV)isanoncytopathic,hepatotropicDNA virusthatcancause chronichepatitis,cirrhosisandhepatocellularcarcinoma 1,2.Viralclearanceduring HBVinfectionistightlyassociatedwiththeappearanceofavigorousvirusspecifi c Tcellresponse3,4.Incontrast,viralpersistenceandchronichepatitisareassociated withamarkedlydiminishedHBVspecificTcellresponse3,4.However,thedifference betweenimmuneresponsespresentinresolvedversuschronicHBVinfectionhas obscuredthediversitypresentwithinchronichepatitisB.ChronicHBVinfectionis ahighlyheterogeneousdisease,andthelevelsofvirusreplication,liverdisease activityandhumoralresponsescandifferconsiderably 5,6 .Basedonbiochemical andvirologicalparametersseveralpotentiallysuccessivephasesofchronicHBV havebeenwelldescribed 7,8 .IntheimmunetolerancephaseserumhepatitisB surfaceantigen(HBsAg)andhepatitisBeantigen(HBeAg)aredetectable;serum HBVDNAlevelsarehighandserumaminotransferasesarenormalorminimally elevated.Intheimmuneclearancephase,serumHBV DNA levelsdecreaseand serumaminotransferaselevelsincrease.Insomepatientsfl aresinthisphase arefollowedbyHBeAgseroconversion. At hirdphase,termedtheinactivecarrier stateischaracterizedbylowlevelsofHBV DNA andnormalconcentrationsof aminotransferases.T reatmentisindicatedforpatientswithactiveinfl ammation andviralreplication.LowinitialHBV DNA levels,high ALTelevationsandHBV genotypehavebeenassociatedwithimprovedresponsetotherapy 911.Treatment isnotindicatedforthoseintheinactivecarrierstate.Interestingly ,thedif ferent profilesofchronichepatitisBhavealsobeenassociatedwithdif ferentmagnitudes ofHBVspecificimmuneresponses.EpisodesofacuteflaresduringchronicHBVare associatedwitharecoveryofHBV specificCD4+ Tc ellresponses 12,13.Increased levelsofHBV specificCD8+ Tc ellresponseshaveonlybeendemonstratedin patientswithalowlevelofHBVreplication,irrespectiveofdegreeofinfl ammation, 1416.These datahaveledtothehypothesesthatineachofthestagesofchronic HBVadistinctintrahepaticimmunologicalmilieuexists,thatdeterminestheresponse totreatment.Thuscharacterizationofthisintrahepaticimmunologicalenvironment isrelevanttobothimprovedunderstandingofchronicHBVinfectionaswellasthe designofantiviraltherapy .The aimofthisstudywastocharacterizethedif ferent phasesofchronicHBVinfectionbyanalysingthecompositionofkeypopulations ofimmuneeffectorcells,i.e.(HBV specific)CD8+Tcells,CD4+TcellsandCD56+ NaturalKiller(NK)cellsintheliver.

Methods

Patients FineNeedleAspiration Biopsies( FNAB)werecollectedfromfi ftynontreated consecutivepatientswithchronicHBVinfectionandstablebiochemicalandvirological parameterswhenmonitoredforaminimumofthreemonthspriortothebiopsy 17,18.

47 CHAPTER4

AllpatientswereHBsAgpositive>6months,HBVDNApositive(>100geq/ml),anti HBVcoreantigen(HBcAg)antibodypositive,andHBeAgorantiHBepositive.Inall patientsantibodiestoHCV,HIV1HIV–2andhepatitisDeltawereabsent.Noneofthe patientshadsignificantcomorbidity.Patientswerestratifiedtothedifferentphases ofchronicHBVaccordingtopredefinedbiochemicalandvirologicalparameters19,20; immunetolerant:alaninetransaminase(ALT)normal(<45IU/l)andHBV DNA>107 geq/ml;immuneclearance:ALT>45IU/landalllevelsofHBV DNA;inactivestate: ALTnormalandHBVDNA≤106 geq/ml.Allpatientssignedinformedconsentbefore thesampleswerecollected.

Virological assessment Bloodsamplesforreportedvirologicalandbiochemicaldatawereobtainedatthe timeoffineneedleaspirationbiopsy.SerumHBeAg,HBsAg,antiHBs,antiHBc, antidelta,antiHCVandantiHIV1/2weredeterminedusingcommerciallyavailable immunoassays(AbbottLaboratories,NorthChicago,IL,USA).SerumHBVDNAwas measuredwithTaqManrealtimePCR(detectionlimit100geq/ml)validatedonthe EurohepStandard21.

PCR and HBV-DNA sequencing HBVDNAwase xtractedfromserumsamplesta kenatthetimeoffi neneedle aspirationbiopsybyusingaMagnaPureLCstation(Roche AppliedScience, Penzberg,Germany).Bothextractionandamplificationefficiencywasmonitoredby spikingthesampleswitha knownamountofcontrolvirus 22.Forsequencingofthe coregene,primerswereusedasdescribedpreviously 23.The amplificationswere purifiedandthecoreregionHBc1827wassequenceddirectlybyusingan ABI 3100automatedsequencer(AppliedBiosystems,Nieuwe kerkaandenI Jssel,the Netherlands).The genotypeofthepredominantHBVpopulationwasdetermined byanalysisofsequencedportionsofthecoreandsurfaceantigenencodingDNA isolatedfromtheserum24. Fine-Needle Aspiration Biopsy Detailsofthefi neneedleaspirationbiopsyprocedurearedescribedelsewhere 25.Brie fly,amandarincontaining25gauge(diameter0.5mm)needle(Braun AG, Melsungen, Germany)ispuncturedinthe8thor9thrightintercostalspace. After removalofthemandarina10mL syringefilledwith2mL RPMIsupplementedwith 0.1%humanserumalbumin(SanquinCLB, Amsterdam,theNetherlands)and25 IU/mL heparin(LeoPharma,Breda,theNetherlands)isattached.Livercellsare aspiratedbynegativesyringepressure.Ne xt,thespecimenistransferredintoa 15mL tubeandplacedoniceimmediatelybeforefurtheranalysis.Validationfor immunologicalassessmentoftheliverusingthefi neneedleaspirationbiopsy aspirate,hasbeendescribedpreviously17,18.

Immunophenotyping Erythrocyteswereremovedfromperipheralbloodandfi neneedleaspiration

48 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

biopsyspecimensbyNH 4CLlysis.The cellsuspensionwaswashedtwicebefore antibodystainingandfourcolorfl owcytometricevaluationusingF ACSCaliber (BectonandDickinsonBiosciences;SanJose,USA(BD))andCELLQuestsoftware (BD).Immunofluorescencestainingwasperformedasdescribedpreviously 16.The followingantibodiesweredividedin2panelsforFACSanalysisofalllymphocytes collectedwiththefineneedleaspirationbiopsy:antiCD4PerCPCy5(BD),antiCD8 FITC(Dako,Glostrup,Denmark(Dako));antiCD8APC(Dako);antiCD56Fitc(BD) andantiCD3PE(Immunotech,Marseille,France).Matchedisotypeantibodieswere usedforcontrolstaining.NKcellsweredefinedasCD8CD56+lymphocytes.

HLA typing, HLA-A2 tetramer staining ScreeningforHLAA2haplotypepositiveHBVpatientswasperformedbystaining PBMCswithanantiHLAA2antibodyderivedfromahybridoma(cloneBB7.2,ATCC HB82).StainingwasfollowedbyanFITCcon jugatedrabbitantimouseantiIgG (Dako)secondaryantibodyandflowcytometricanalysis.InHLAA2+HBVpatients HBVspecificTcellswerestainedwiththesolubleHLAA2peptidetetramer:HBc18 27(FLPSDFFPSV)APCconjugated(ProImmune,Oxford,UK)togetherwithantiCD8 FITC(Dako).Stainingwasperformedasreportedbefore 16.Incontrole xperiments withPBMCsofHLAA2positivehealthycontrols(n =4)andHLAA2negativeHBV patients(n=4)thetetramerboundto0.019±0.002and0.013±0.004%oftotalCD8+ Tc ellsrespectively.Therefore backgroundstaininglevelwasdetermined 0.025% (mean+2SD).

Statistical analysis Dataaree xpressedasmean ±SEM,unlessindicatedotherwise.Onewayand multivariableanalysisofvariancewasperformedwithSPSS1 1.0.1for Windows (SPSS,Chicago,IL).ForpairwisecomparisonBonferronicorrectionwasapplied. Statisticalanalyseswereperformedusing χ2forcategoricalvariables.Correlations weredeterminedusingtheSpearman ’scorrelationtest.Inallanaly zesapvalueof <0.05wasconsideredstatisticallysignificant.

Results

Patients Of50consecutivechronicHBVpatientssampled,4 7wereeligible. Threepatients wereexcludedfromfurtheranalysis:onepatientduetocoexistingalcoholabuse,one forreceivinglamivudinetherapy,andoneasaconse quenceofnonrepresentative fin eneedleaspirationbiopsyincytospinevaluation. The47chronicHBVpatients, 36(72%)maleandwithamedianageof35(1469),werestratifiedtothedifferent phasesofchronicHBVinfectionbasedonpredefi nedbiochemicalandvirological status(tableI).Sevenpatientswereassignedtotheimmunetolerantphasewitha medianALTof33IU/l(1643),medianviralloadof2.5x109geq/ml(1.1x1071.6x1010) andallHBeAgpositive.Twentyfourpatientswereassignedtotheimmuneclearance

49 CHAPTER4 phasewithamedianALT of91.5IU/l(48863),medianviralloadof9x10 7 geq/ml (1.6x1031.1x1010)and10HBeAgpositive.Sixteenpatientswereinactivecarriers withmedianALT of25.5IU/l(1645),medianHBV DNA of3.4x10 3 geq/ml(162 1.2x106),allHBeAgnegative.Exceptforrace,therewasevendistributionofage,sex andgenotype.Themeannumberofcellsobtainedbyfi neneedleaspirationbiopsy andanalyzedbyflowcytometrywas7316(±1463)forpatientsintheimmunetolerant phase,8159(±232)forpatientsintheimmuneclearancephase,and7349(±459)for inactivecarriers.Thedifferencesinthesecellcountswerenotsignificant.Allsamples werecollectedwithoutanycomplicationstothepatients.

Table I. Patient characteristics at the time of aspiration biopsy.

Patients Immune Immune Inactive n=47 tolerance clearance Carriers (n=7) (n=24) (n=16) ∗ Age 41 31.5 35.5 (19-69) (16-62) (17-51) Sex 3 M, 4 F 20 M, 4 F 10 M, 6 F Race+ 4 Asian, 1 Cauc., 5 Asian, 17 Cauc., 7 Asian, 5 Cauc., 2 Mixed Race 2 Mixed Race 4 Mixed Race ∗ ALT 33 91.5 25.5 (U/l) (16-43) (48-863) (16-45) ∗ 9 7 3 HBV-DNA 2.5x10 9x10 3.4x10 7 10 3 10 6 (geq/ml) (1.1x10 -1.6x10 ) (1.6x10 -1.1x10 ) (162-1.2x10 ) HBeAg 7 10 0 positive Genotype 2-0-4-0-1 4-4-2-11-3 3-2-5-2-3 1ND A-B-C-D-E Cirrhosis - 3x 1x

∗ Median values (range). ND: Not Determined + Uneven distribution; p = 0.037 in χ2 test.

ThecompositionofliverderivedlymphocytesinpatientswithchronicHBVinfection CD3CD56+NKcells,CD4+ TcellsandCD8+ Tc ellshaveallbeenshowntobe involvedintheantiHBVimmuneresponse 13,14,26.Theproportionoftheseimmune effectorcellsinliverderivedlymphocytesandPBMCsofpatientswithchronicHBV issummarizedinfigure1.InimmunetolerantpatientsthefractionNKcellsinthe liverwashigherthaninpatientsintheimmuneclearanceandinactivephase;mean percentage41.1±2.8,29.8±1.7and35.0±2.0respectively(immunetolerancevs. immuneclearancep=0.006,immunetolerantvs.inactivecarriersp=0.052).Alsoin PBMCsthefractionNKcellswashigherinimmunetolerantpatientsthaninpatients intheimmuneclearanceandinactivephaseofchronicHBV;mean%28.2(±4.5), 17.1(±1.8)and18.7(±2.9)respectively(immunetolerancevs.immuneclearancep

50 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

=0.015,immunetolerantvs.inactivecarriersp=0.047). NodifferenceswerefoundwithrespecttotheproportionCD4+ TcellsorCD8+ T cellsinthedifferentphasesofchronicHBV,eitherintheliverorperipheralblood.In patientsintheimmunetolerantphase,theimmuneclearancephaseandtheinactive carrierstatethemeanpercentageCD4+Tcellsintheliverwas25.2±2.8,25.8±1.7 and22.8±2.0,respectively,inperipheralblood37.7(±2.5)41.6(±2.0)and39.7(± 2.3)respectively.InallthreephasesthelivercontainedmoreCD8+TcellsthanCD4+ Tc ells,whereasthiswastheoppositeinperipheralblood.Themeanpercentage CD8+Tcellsintheliverwas34.5±2.7inpatientsintheimmunetolerantphase,36.7 ±2.0inpatientsintheimmuneclearancephaseand37.9±2.2ininactivecarriers.In peripheralbloodthesepercentageswereconsiderablylower;29.7(±3.1),29.3and (±1.9)31(±1.9)respectively.Resultswereindependentofage,sex,raceandHBV genotype.

Liver Peripheral blood * +

50 50 * + 40 40 e g a t 30

n 30 e c r

e 20 20 P

10 10

0 0 CD8+ cells CD4+ cells CD56+ cells CD8+ cells CD4+ cells CD56+ cells Immune effector cell Immune effector cell

* p = 0.006 Immune tolerance Immune clearance Inactive carriers * p = 0.015 + p = 0.052 + p = 0.047

Figure1. MeanpercentageCD8+Tcells,CD4+TcellsandNKcellsinliverderivedlymphocytes andperipheralbloodofpatientsintheimmunetolerantphase(),patientsinthe immuneclearancephase()andinactivecarriers(). IntheliverandperipheralbloodofimmunetolerantpatientsNKcellsarethedominant lymphocytepopulation.IntheliverofallthreepatientgroupsthefrequenciesofCD8+ Tcellsaredominantoverthefre quenciesofCD4+ Tcells,whereastheoppositeis thecaseinperipheralblood.

The composition of peripheral blood and liver derived lymphocytes in patients in the immune-clearance phase of chronic HBV IntheimmuneclearancephaseserumHBVDNAlevelsvariedfromhightolowand someindividualshadseroconvertedfromHBeAgpositivetoanti–HBeAgpositive. Recently,itwassuggestedthataserumHBV DNAof107geq/mlappearsadividing thresholdbelowwhichcirculatingHBVspecificTcellscanconsistentlybedetectedex vivo 14,suggestingthatthedifferentvirologicalstagesmaybetheresultofdynamics intheimmuneresponse.Therefore,thecompositionofliverderivedlymphocytesin

51 CHAPTER4

patientswithhighviralload(HBVDNA>107geq/ml;HBVhigh)wascomparedtoliver derivedlymphocytesinpatientswithlowviralload(HBVDNA<107geq/ml;HBVlow). TableIIsummarizesthecharacteristicsofthe2patientgroups.Of24patientsinthe immuneclearancephase14hadhighviralload,withamedianHBVDNAof5.7x108 geq/ml(2x1071.1x1010)andamedianALTof95IU/L(48863).Tenpatientshadalow virustiterwithamedianHBVDNAof1.9x104geq/ml(1.6x1033.5x106)andamedian serumALTof89IU/L (63120).Thediff erenceinALT levelsbetweenthe2groups wasnotsignificant.Of14HBVhighpatients10wereHBeAgpositive,whereasall HBVlowpatientshadseroconvertedtoantiHBepositive.

Table II: Patients in the immune-clearance phase

Immune-clearance HBV-DNA HBV-DNA Patients >1x107 geq/ml <1x107 geq/ml (n=24) (n=14) (n=10) ∗ Age 30 36.5 (20-62) (16-59)

Sex 10 M, 4 F 10 M Race+ 2 Asian, 3 Asian, 5 Cauc., 12 Cauc. 2 Mixed Race ∗ ALT 95.5 89 (U/l) (48-863) (63-120) ∗ 8 4 HBV-DNA 5.7x10 1.9x10 7 10 3 6 (geq/ml) (2x10 -1.1x10 ) (1.6x10 -3.5x10 ) HBeAg 10 0 positive Genotype 2-2-1-9-0 2-2-1-2-3 A-B-C-D-E ∗ Median values (range). + Uneven distribution; p < 0.05 in χ2 test.

The difference in serum ALT levels between IC-hi and IC-lo was not significant.

TheproportionofCD4+ Tcells,CD8+TcellsandNKcellsintheliverofpatientsin theimmuneclearancephaseissummarizedinfi gure2.Comparedtopatientswith highHBVDNA,intheliverofthosewithlowHBV DNAthemeanpercentageCD4+ Tc ellswashigher;22.5%(±1.8)vs.30.4%(±1.4)respectively(p=0.02). This differencewasnotfoundinPBMCs;themeanpercentageCD4+ Tcellswas41.2(± 3.1)and42.1(±2.1)respectively.Interestingly,theliverofpatientsintheimmune clearancephaseandlowviralloadcontainedasignificantlyhigherproportionCD4+ Tcellsthaninactivecarriers(30.4%±1.4vs.22.8±2.0,respectively,p=0.02).In

52 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

contrast,inimmuneclearancepatientswithhighviralloadthefractionCD8+ Tcells washigherthaninpatientswithlowviralload(meanpercentage40.3±2.4and31.7 ±2.8respectively(p=0.02)).Again,thisdifferencewasabsentinperipheralblood; 31.4(±2.8)vs.26.3(±2.2),respectively. Nodifferencewasobservedbetweenthetwoimmuneclearancegroupsregarding theproportionofNKcellsintheliverandperipheralblood.Intheliverofpatientswith highviralload29.5%±2.5oftotallymphocyteswereNKcells,ascomparedto30.2% ±2.2inpatientswithlowviralload.Inperipheralbloodthemeanswere15.2%(±2.1) and19.6%(±3.0)respectively. To excludeassociationbetweenproportionsofintrahepaticlymphocytepopulations aspearmancorrelationtestwasperformed.Onlyweaknegativecorrelationswere foundbetweenCD8+TcellsandCD4+Tcells(r=0.3),betweenCD4+Tcellsand NKcells(r=0.2)andbetweenCD8+TcellsandNKcells(r=0.2).Theintrahepatic distributionofallinvestigatedlymphocytesubsetswasindependentofage,sex, genotypeandrace.

Liver Peripheral Blood

50 p < 0.05 50

40 40 e g a t 30 30 n e c r

e 20 20 P

10 10

0 0 CD8+ cells CD4+ cells CD56+ cells CD8+ cells CD4+ cells CD56+ cells Immune effector cell Immune effector cell

High viral load Low viral load Figure2. MeanpercentageCD8+Tcells,CD4+TcellsandNKcellsinliverderivedlymphocytes andPBMCsofpatientsintheimmuneclearancephase.Patientsweredividedbased uponHBVDNAtiters;>107geq/ml()and<107geq/ml(). IntheliverofimmuneclearancepatientswithhighviralloadtheproportionCD8+ T cellswassignificantlyhigherthaninpatientswithlowviralload.Intheliverimmune clearancepatientswithlowviralloadthepercentageCD4+ Tcellswassignificantly higherthaninpatientswithhighviralload. Thesedif ferenceswereabsentin peripheralblood.

Ex Vivo Frequency of HBVspecificCD8+Tcellsinliver Fifteenof47patients(32%)wereHLAA2positive;3patientsintheimmunetolerant phase,9intheimmuneclearancephaseand3inactivecarriers(tableIII).Inthese casesstainingofHBVspecificTcellswasperformedwithsolubleHLAA2tetrameric complexesabletovisualize Tcellsspecific forthecoreepitope1827.In3of15 patientstetramerstainingexceededthebackgroundlevelof0.025%oftotalCD8+

53 CHAPTER4

Tcellpopulation.All3wereintheimmuneclearancephase;1patientwithhigh viralload(case4)and2withlowviralload(cases10and12)(fi g3).Interestingly, intheonlypatientwithhighHBV DNA anddetectableHBVspecifi c CD8+Tcells (case4),serumALT of187IU/LnormalizedandHBV DNAdecreasedfrom2x10 7 geq/mlto1x10 4geq/mlwithinamonthaftersampling.Theabsenceofcore1827 specificCD8+Tcellsmaybecausedbyviralmutationsresultinginspecifi cepitope inactivation14.Therefore,inall15HLAA2+patientsthiscoreepitopewassequenced. SixpatientswereinfectedwithanHBVstraincarryingavalinetoisoleucinemutation atposition27.However,thisaminoacidmutationisnotassociatedwithcore1827 unresponsiveness,sinceitwasalsodetectedinapatientwhoshowedaCD8+Tcell responseagainstthisepitope.Oneinactivecarrierinwhomnotetramerpositivecells weredetected(case15),displayedanAsparticacid–to–Asparaginesubstitutionat position22,whichisassociatedwithreducedCD8+Tcellactivation27. s l l 3 e

A c 2.6

+

8 2.5 D C l

a 2 t o T / 1.5 1.3 Peripheral Blood +

8 Liver D

C 1

+ . t

e 0.5 T 0.19 0.22 0.02 0.02 % 0 Case 4 Case 10 Case 12 HBV DNA (geq/ml) 2x107 1.5x104 2.4x104 ALT (IU/l) 187 85 111 HBeAg negative negative negative

B HLA A2+ HBV patient (case 4) HLA-A2 neg HBV patient Liver Periphereral blood Peripheral blood

C 1.3% 0.19% 0.01% P A

7 2 - 8 1 c B H

CD8 Fitc Figure3.HBc1827tetramerbindingtoCD8+Tcells. 3a)PercentageHBc1827tetramerpositiveCD8positive Tc ellsintheliverand inperipheralbloodofthe3patientsinwhichthesecellsweredetected(Liver, Peripheralblood).Inall3patientsthereissequesteringoftetramerpositivecells intheliver.IntheonlypatientwithhighHBVDNAanddetectableHBVspecificCD8+ Tcells,serum ALTnormalizedandHBV DNA decreasedto1x10 4geq/mlwithina monthaftersampling. 3b)PercentagesHBc1827tetramer+CD8+ Tc ellsinarepresentativeHLAA2+ patient(case4). Thenumberintheupperrightquadrantindicatesthepercentage HBc1827tetramer+CD8+TcellsoftotalCD8+Tcells.Therightpanelshowsdata ofarepresentativeHLAA2chronicHBVpatient,servingascontrolforaspecifi c tetramerbinding.

54 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

Discussion

ItisnowwidelyacceptedthatchronicHBVpatientscanbegroupedintodifferentwell definedphases:aninitialreplicative,immunetolerancephase,followedbygradual decreaseofviralreplicationintheimmuneclearancephaseandathirdasymptomatic inactivecarrierstate7,8. Wefoundthatinimmunetolerantpatientsbutnotinimmuneclearancepatients andinactivecarriers,lymphocytesthatbeartheclassicalNKphenotypearethe dominantimmuneef fectorcellpopulationintheliver .Infact,thedistributionofNK cells,CD4+andCD8+ Tc ellsinimmunetolerantpatientsresemblesthesituation observedpreviouslyinthenormalhumanliver 28whichmaysupporttheassumption thatimmunetolerantpatients,characterizedbyhighviralloadandnormalALTlevels havealowendogenousimmuneresponsetoHBV9,29. Incontrasttoimmunohistochemicalstainingofastandardliverbiopsy,thefineneedle aspirationbiopsydoesnotallowthenecessaryequalsamplesizeineachpatientin ordertocompareabsolutecellnumbers.Itisknownthatinactivecarriershaveless cellularinfiltratethanpatientswithactivehepatitis 16andthereforetheabsolute numberofNKcellspresentintheliverofimmunetolerantpatientsmaybelowerthan intheimmuneclearancephase. However, similartopreviousimmunohistochemistrystudieswefoundarelative deficiencyofCD4+ Tc ellscomparedtoCD8+ Tc ellsintheliverofpatientswith chronicHBV 30,31.Interestingly,inthesepreviousstudiesitwasnoticedthatinthe HBeAgnegativesubpopulationoftheindividualswithactivehepatitis,thefraction CD4+T cellshadincreasedcomparedtoHBeAgpositivepatientswithactive hepatitis30.Indeed,wefoundthatintheimmuneclearancephase,thepatientswith highviralloadofwhichalargemajorityisHBeAgpositive,hadalowerproportionof intrahepaticCD4+Tcells,thanpatientswithlowviralloadofwhichnonewereHBeAg positive.Theimmuneclearancephasecomprisesaheterogenicpatientpopulation, andthevariousvirologicalstagesmaybetheresultofdynamicsintheintrahepatic immuneresponse. Alternatively,theincreasedfractionofintrahepaticCD4+ Tcellsinpatientswithlow HBVDNAandhepatitismightbeaconsequenceandnotthecauseoflowHBVDNA. Nevertheless,apreviousstudyhasshownthatCD4+ Tcellsarerequiredtosustain CD8+Tcellresponsesduringchronicviralinfection 32.Therefore, anincrementin thefractionCD4+Tcellsintheliverofpatientsintheimmuneclearancephase,may contributetoanimprovementoftheHBV specificCD8+Tcellresponsewhichmay resultinadecreasingviralload.Ourobservationandthatbyothers 1416,thatHBc 1827–specificCD8+ Tc ellsareprimarilyfoundintheliverofpatientswithactive hepatitisandlowHBVDNA,maysupportthisconcept.Unexpectedly ,inonepatient intheimmuneclearancephasewithahighviralload,wefoundhighfrequenciesof intrahepaticHBc1827specificCD8+cells.Shortlyafterobtainingtheliversample, HBVDNAlevelsdroppedfrom2x107geq/mlto1x104geq/ml.Althoughthismayhave beenthetimepointatwhichtheantiviralimmuneresponsewasdeveloping,additional

55 CHAPTER4

sampleswerelackingforsequentialanalysisandfollowup. InchronicHBVandchronichepatitisCinfectiontheeffi cacyofinterferonalphain inducingsustainedvirologicresponsehasbeenattributedtotheefficacyininduction andmaintenanceofsignificantCD4+Tcellresponses 3334.Theimprovedresponse toantiviraltherapyofchronicHBVpatientswithhepaticinfl ammationandlowviral loadcomparedtopatientswithinflammationandhighviralload 9,10maybeadirect consequenceofanincrementinintrahepaticCD4+Tcells. ThedifferencesinproportionCD8+ Tc ellsandCD4+ Tc ellswereabsentin peripheralblood,whichunderlinesthevalueofstudyingintrahepaticimmunology ascomparedtoimmuneef fectorcellsderivedfromthecirculation,ininvestigating immunepathologyinchronicHBVinfection. Inconclusion,analysisofthephenotypeofintrahepaticimmuneresponsesuggests thattheclinicalphasesofchronicHBVinfection,separatedonbiochemicaland virologicalparameters,havecharacteristicintrahepaticimmuneresponses.In immunetolerantpatientsNKcellsarethedominantimmuneef fectorcellpopulation intheliver,andintheimmuneclearancephasethepatientswithlowviralloadhad ahigherproportionofintrahepaticCD4+ Tcells,thanpatientswithhighviralload. Thesefindingsarerelevanttothedesignofnew,individualisedantiviralstrategies.

56 CHAPTER4CHAPTER4IMMUNETOLERANCEVS.IMMUNECLEARANCE

References 1. KaneM.GlobalprogrammeforcontrolofhepatitisBinfection.Vaccine1995;13Suppl1:S479. 2. WrightTL,LauJY.ClinicalaspectsofhepatitisBvirusinfection.Lancet1993;342:13404. 3. FerrariC,PennaA,Bertoletti A,etal.CellularimmuneresponsetohepatitisBvirusencodedantigens in acuteandchronichepatitisBvirusinfection.JImmunol1990;145:34429. 4. JungMC,SpenglerU,SchrautW,etal.HepatitisBvirusantigenspecificTcellactivationinpatientswith acuteandchronichepatitisB.JHepatol1991;13:3107. 5. LokAS,McMahonBJ.ChronichepatitisB.Hepatology2001;34:122541. 6. MaruyamaT,McLachlanA,IinoS,KoikeK,KurokawaK,MilichDR.TheserologyofchronichepatitisB infectionrevisited.JClinInvest1993;91:258695. 7. deFranchisR,HadengueA,LauG,etal.EASLInternationalConsensusConferenceonHepatitisB.1314 September,2002Geneva,Switzerland.Consensusstatement(longversion).JHepatol2003;39Suppl1: S325. 8. LokAS.ChronichepatitisB.NEnglJMed2002;346:16823. 9. LokAS, HeathcoteEJ,HoofnagleJH.ManagementofhepatitisB:2000summaryofaworkshop. Gastroenterology2001;120:182853. 10. SchalmSW,HeathcoteJ,CianciaraJ,etal.Lamivudineandalphainterferoncombinationtreatmentof patientswithchronichepatitisBinfection:arandomisedtrial.Gut2000;46:5628. 11. JanssenHL,vanZonneveldM,SenturkH,etal.Pegylatedinterferonalfa2baloneorincombinationwith lamivudineforHBeAgpositivechronichepatitisB:arandomisedtrial.Lancet2005;365:1239. 12. TsaiSL,ChenPJ,LaiMY ,etal.AcuteexacerbationsofchronictypeBhepatitisareaccompaniedby increasedT cellresponsestohepatitisBcoreandeantigens.ImplicationsforhepatitisBeantigen seroconversion.JClinInvest1992;89:8796. 13. RossolS,MarinosG,CarucciP ,SingerMV ,WilliamsR,NaoumovNV.Interleukin12inductionof Th1 cytokinesisimportantforviralclearanceinchronichepatitisB.JClinInvest1997;99:302533. 14. WebsterGJ,ReignatS,BrownD,etal.LongitudinalanalysisofCD8+ Tcellsspecificforstructuraland nonstructuralhepatitisBvirusproteinsinpatientswithchronichepatitisB:implicationsforimmunotherapy. JVirol2004;78:570719. 15. SobaoY,TomiyamaH,SugiK,etal.TheroleofhepatitisBvirusspecificmemoryCD8Tcellsinthecontrol ofviralreplication.JHepatol2002;36:10515. 16. MainiMK,BoniC,LeeCK,etal.Theroleofvirusspecifi cCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 17. SprengersD,vanderMolenRG,KustersJG,etal.Flowcytometryoffi neneedleaspirationbiopsies:a newmethodtomonitortheintrahepaticimmunologicalenvironmentinchronicviralhepatitis.JViralHepat 2005;12:50712. 18. SprengersD,vanderMolenRG,KustersJG,JanssenHL.Coarsevs.fi neneedleaspirationbiopsy .J Hepatol2004;41:5034;authorreply504. 19. ChuCJ,HussainM,LokAS.QuantitativeserumHBVDNAlevelsduringdifferentstagesofchronichepatitis Binfection.Hepatology2002;36:140815. 20. HadziyannisSJ,VassilopoulosD.HepatitisBeantigennegativechronichepatitisB.Hepatology2001;34: 61724. 21. PasSD,FriesE,DeManRA,OsterhausAD,NiestersHG.Developmentofaquantitativerealtime detectionassayforhepatitisBvirusDNA andcomparisonwithtwocommercialassays.JClinMicrobiol 2000;38:2897901. 22. NiestersHG.Clinicalvirologyinrealtime.JClinVirol2002;25Suppl3:S312. 23. StuyverL,DeGendtS,V anGeytC,etal. An ewgenotypeofhepatitisBvirus:completegenomeand phylogeneticrelatedness.JGenVirol2000;81:6774. 24. NorderH,Courouce AM,MagniusLO.Completegenomes,phylogeneticrelatedness,andstructural proteinsofsixstrainsofthehepatitisBvirus,fourofwhichrepresenttwonewgenotypes.V irology1994; 198:489503. 25. LautenschlagerI,HockerstedtK,HayryP.Fineneedleaspirationbiopsyinthemonitoringofliverallografts. TransplInt1991;4:5461. 26. KakimiK,GuidottiLG,Koezuka Y, ChisariFV.Naturalkiller Tc ellactivationinhibitshepatitisBvirus replicationinvivo.JExpMed2000;192:92130. 27. BertolettiA,SouthwoodS,ChesnutR,etal.MolecularfeaturesofthehepatitisBvirusnucleocapsidTcell epitope1827:interactionwithHLAandTcellreceptor.Hepatology1997;26:102734. 28. NorrisS,CollinsC,DohertyDG,etal.Residenthumanhepaticlymphocytesarephenotypicallydif ferent fromcirculatinglymphocytes.JHepatol1998;28:8490. 29. LauGK,NanjiA,HouJ,etal.Thymosinalpha1andfamciclovircombinationtherapyactivates Tcell

57 CHAPTER4

responseinpatientswithchronichepatitisBvirusinfectioninimmunetolerantphase.JViralHepat2002; 9:2807. 30. MontanoL,AranguibelF,BoffillM,GoodallAH,JanossyG,ThomasHC.Ananalysisofthecompositionof theinflammatoryinfiltrateinautoimmuneandhepatitisBvirusinducedchronicliverdisease.Hepatology 1983;3:2926. 31. YangPM,SuIJ,LaiMY,etal.Immunohistochemicalstudiesonintrahepaticlymphocyteinfiltratesinchronic typeBhepatitis,withspecialemphasisontheactivationstatusofthelymphocytes.AmJGastroenterol 1988;83:94853. 32. MatloubianM,ConcepcionRJ, AhmedR.CD4+ Tc ellsarerequiredtosustainCD8+cytotoxic Tcell responsesduringchronicviralinfection.JVirol1994;68:805663. 33. MarinosG, TorreF,ChokshiS,etal.Inductionof ThelpercellresponsetohepatitisBcoreantigenin chronichepatitisB:amajorfactorinactivationofthehostimmuneresponsetothehepatitisBvirus. Hepatology1995;22:10409. 34. KamalSM,FehrJ,RoeslerB,Peters T, RasenackJW.Peginterferonaloneorwithribavirinenhances HCVspecificCD4Thelper1responsesinpatientswithchronichepatitisC.Gastroenterology2002;123: 107083.

58 CHAPTER 5

Analysis of intrahepatic HBV-specifi c cytotoxic T-cells during and after acute HBV infection in humans

D.Sprengers1,R.G.vanderMolen1,J.G.Kusters1,R.A.DeMan1,H.G.M.Niesters2, S.W.Schalm1,H.L.A.Janssen1.

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

JHepatol2006;45:182189 CHAPTER5

Abstract

CharacteristicsoftheintrahepaticvirusspecificTcellresponseinpatientswithacute hepatitisBvirus(HBV)infectionhavenotbeenstudiedduetotheriskofcomplications associatedwithstandardliverbiopsies.Inthisstudyweaimedtocharacterizethe virusspecificCD8+TcellresponseintheliverofpatientswithacuteHBVinfection usingthefineneedleaspirationbiopsy(FNAB). InHLAA2positivepatientswithacuteHBVinfectionaFNABwasdoneatfi rst presentation,atthetimeofHBsAgseroconversionandthreemonthsafterHBsAg seroconversion.HLAA2tetramerswereusedtoidentifyHBV specificCD8+Tcells inFNABcytologyandperipheralblood(PB). Atfirstpresentationtherewasacorrelationbetweenthefrequencyofintrahepatic CD8+Tcellsandthedegreeofliverdamage.Atalltimepointstherewassequestering ofHBVspecificCD8+Tcellsintheliver,andthepercentageofintrahepaticHLADR expressingHBVspecificCD8+ TcellswashigherthaninPB. Threemonthsafter HBsAgseroconversionthefrequencyofintrahepaticHBV specificCD8+ Tcells remainedhigh. Inconclusion,HBVspecificCD8+ Tcellsarecompartmentalizedintheliverduring acuteHBVinfection.Theirpresenceinthelivermaysuggestaroleintheresolution oftheinfection.IntrahepaticHBV specificCD8+ Tc ellsremaindetectableathigh frequenciesafterHBsAgseroconversion.

60 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

Introduction

CytotoxicTlymphocytes(CTL)contributetocontrolofhepatitisBvirus(HBV)infection byinhibitingviralreplicationininfectedlivercells13.Patientswithacutehepatitiswho ultimatelycontrolthevirus,possessastrongCTLresponseinperipheralblood(PB) thatisabletorecognizedifferentepitopeswithinHBVproteinsandpersistswithout liverdamage24.Incontrast,thisCTLresponseisbarelydetectableinthecirculation ofchronicpatientsdespitebiochemicalandhistologicalsignsofliverdamage 5 6. InhumanHBVinfection,studieshavebeenhamperedbythediffi cultyofstudying theintrahepaticcompartment. ThefewstudiesthathaveinvestigatedHBV specific immuneresponsesintheliverhavefocussedonchronicinfectionandshowed importantdifferencesbetweencirculatingandintrahepaticHBVspecificCD8+Tcells withrespecttophenotype,activationstatusandfrequency78.ThevirusspecificTcell responseatthesiteofviralreplicationhasnotbeenstudiedyetinacuteHBVpatients. Inthissetting,samplingoftheliverisrarelyjustifiableduetotheseriouscomplications associatedwithstandardlivertissuebiopsytechniques 9.Thefin eneedleaspiration biopsy(FNAB)oftheliverwithaninvasivenesscomparabletoavenapunctionis muchbettertolerated,andcanthereforeberepeatednumeroustimes 10,1 1.W e haverecentlyshownthatfl owcytometricanalysisofliverderivedlymphocytesin theFNABaspirateofpatientswithchronicviralhepatitis,allowsreliableanalysis oftheintrahepaticcompartment 10,12 .This observationhasfacilitatedthepresent studyinwhichweaimedtocharacterizethevirusspecifi cCD8+Tcellresponsein theliverofpatientswithacuteHBVinfection.HLAA2tetramericcomplexeswith3 differentpeptidesrepresentingHBVcore,envelopeandpolymeraseepitopeswere usedtoidentifyHBVspecificCD8+TcellsinFNABcytology.Theseantigenspecific populationswerefollowedoverthecourseofinfectionandtheirphenotypeand frequencieswerecorrelatedwithbiochemicalparameters.

Methods

Patients BetweenJanuaryandJuly2004ninepatientswithacuteHBVinfectionwerereferred toourclinicbylocalCommunityHealthCenters( GGD).FiveHLAA2positiveadult subjectswithselflimitedacuteHBVinfectionparticipatedinthepresentstudyonthe basisoftheexpressionoftheHLAA2alleleandinallfivepatientsthevirusspecific CD8+Tcellresponseintheliverandperipheralbloodwasmonitoredatthetimeof fir stpresentation(day0)andsoonafterHBsAghaddisappearedfromthecirculation (HBsAgseroconversion).Infourpatients(patient1,2,3and5)anadditionalsample wascollectedatday30and/or90dayspostHBsseroconversion. Diagnosisofacutehepatitiswasbasedonincreasedserumalanineaminotransferase (ALT)activity(atleast10timestheupperlimitofnormal),fi rstdetectionofhepatitis Bsurface(HBsAg)antigenandimmunoglobulin(Ig) MantihepatitisBcoreantigen (antiHBc)antibodiesintheserum,alongwiththerecentonsetofjaundiceand

61 CHAPTER5

associatedsymptoms.InallpatientsantibodiestohepatitisCvirus(HCV),HIV 1HIV–2andhepatitisDeltawereabsent.Noneofthepatientshadsignifi cantco morbidity.Allpatientsrecoveredclinically;thetransaminaselevelsnormalized,and HBsAgandHBeAgantigensbecamenegativeintheserum.Allpatientsgavewritten informedconsentbeforethesampleswerecollected. Thisstudywasperformedin agreementwiththelocalMedicalEthicsCommittee.

FNABprocedure Intrahepaticimmunecellswereobtainedbyfi neneedleaspirationbiopsy( FNAB). Detailsofthe FNABprocedurearedescribedelsewhere 1012.Brie fly,amandarin containing25gauge(diameter 0.5mm)needle(Braun AG,Melsungen, Germany) ispuncturedinthe8thor9thrightintercostalspace.Afterremovalofthemandarina 10mLsyringefilledwith2mLRPMIsupplementedwith0.1%humanserumalbumin (SanquinCLB,Amsterdam,theNetherlands)and25IU /mLheparin(LeoPharma, Breda,theNetherlands)isattached.Livercellsareaspiratedbynegativesyringe pressure.Next,theneedleisflushedwith3mLculturemediumtoobtainthelivercells frominsidetheneedleandthespecimenistransferredintoa15mLtubeandplaced oniceimmediately.

Virologicalassessment Bloodsamplesforvirologicalandbiochemicaldatawereobtainedatthetimeof FNAB.SerumHBsAg,antiHBsAg,HBeAg,antiHBcAg,antidelta,antiHCVand antiHIV1/2weredeterminedusingcommerciallyavailableimmunoassays(Abbott Laboratories,NorthChicago,IL,USA).SerumHBVDNAwasmeasuredwithTaqMan realtimePCR(detectionlimit362geq/ml)validatedontheEurohepStandard13.

PCRandHBVDNAsequencing HBVDNAwasextractedfromserumsamplesta kenatthetimeof FNABbyusinga MagnaPureLCstation(RocheAppliedScience,Penzberg,Germany).Bothextraction andamplificationefficiencywasmonitoredbyspi kingthesampleswitha known amountofcontrolvirus14.Forsequencingprimerswereusedasdescribedpreviously 15.The amplificationswerepurifiedandthecoreregion1 827,envelope1 83191 andpolymerase816824weresequenceddirectlybyusingan ABI3100automated sequencer(AppliedBiosystems,NieuwerkerkaandenIJssel,theNetherlands).The genotypeoftheHBVpopulationwasdeterminedbyanalysisofse quencedportions ofthecoreandsurfaceantigenencodingDNAisolatedfromtheserum16.

HLAtyping PBMCwereisolatedfromheparinizedbloodsamplesbydensitygradientcentrifugation onFicollHypaque(AmershamPharmaciaBiotechAB,Uppsala,Sweden).Screening forHLAA2haplotypepositivitywasperformedbystainingPBMCwithanantiHLA A2antibodyderivedfromahybridoma(cloneBB 7.2,ATCCHB 82).Stainingwas followedbyanFITCconjugatedrabbitantimouseantiIgG(Dako,Glostrup,Denmark

62 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

(Dako)secondaryantibodyandflowcytometricanalysis.

ImmunophenotypingandHLAA2tetramerstaining

Erythrocyteswereremo ved from FNABspecimensbyNH 4CLlysis.T hiscell suspensionandisolatedPB MCwerewas hedtwicebe foreantibodystainin g and fourcolorfl owcytometrice valuation using FACSCaliber(Bectonand DickinsonBiosciences ;San Jose,USA( BD))andCELL Questso ftware(BD). Immunofluorescencestainin gwas performedasdescribed previously 4.HBV specificTcellswerestainedwit hsolubleHLAA2/peptidetetramers.Se quencesof HBV(genotypeD)peptidesusedwere:FLPSDFFPSV(core1827)PEconjugated, FLLTRILTI(envelope183191)APCconjugated,andSLYADSPSV(polymerase816 824)APCconjugated(allmanufacturedbyProImmune,Oxford,UK).Thesesoluble HLAA2/peptidetetramersincombinationwit hantiCD4PE(BD),antiCD8 FITC (Dako)andantiHLADRPerCP (BD)monoclonalantibodies( MAbs)weredi vided in2 panelsforFACSanalysiso falllymphocytescollectedby FNAB,andatleast 2x105PBMCperpanel.Matchedisotypeantibodieswere usedforcontrolstainin g. Incontrole xperimentswithPBMCand FNABcytologyofHLAA2positivepatients withchronicnonHBVrelatedliverdisease(n=4)andHLAA2negativechronicHBV patients(n=4)thelevelofaspecific tetramerbindinginPBMCwasdeterminedas 0.03%oftotalCD8+Tcells(mean +2SD).In FNABcytologythispercentagewas alwayslower.InanalysisoftetramerbindinginPBMCandliverderivedlymphocytes backgroundstaininglevelwasseton0.03%oftotalCD8+Tcells.

Results

Patients Figure1showsthepatientcharacteristicsandthedynamicsofHBVreplicationand liverinflammation,asevidencedbyserumALTlevels,atthetimepointsofsampling. All patientss howedasimilar patterno fserumHBVand ALTreduction.At first presentation,highlevelsofserumALTandHBVDNAcoexisted.Bothparameters rapidlydeclinedandser umALThadreachednormallevelsbythetimethatHBsAg wascleared fromt hecirc ulation.All patients presentedato uro utpatientclinic aftertheonseto fsymptomsand proceededtoclinicalreco verywithouttherapeutic interventions.

RatioofCD8+andCD4+cellsduringacuteHBVinfection To investigatet hedynamicso fbothlymphocytes ubsetsin humanac uteHBV infection,atday 0andatt hetimeo fHBsAgseroconversionwedeterminedt he percentageso fcirculatingandintra hepaticCD 8+andCD 4+cellswit hint hetotal lymphocytepopulation(mean±SEM).AtbothtimepointsthepercentageofCD8+T cellswashigherintheliver(47.6±1.6and36.9±2.4,respectively)thaninPB(30.5 ±2.0and23.1±1.5,respectively),andthepercentageofCD4+Tcellswashigher inthecirculation(38.2±5.1and50.5±1.5,respectively)thanintheliver(21.1±3.1

63

CHAPTER5

T e t r +

C D 8 +

c e l l s /

t o t a l C D 8 +

c e l l s ( %

)

L

)

l

D

5

5

5

.

T

,

,

,

m

t

/

c

0

0

1

1

2

2

s

+

+

q

o

o

r

8

8

e

p

e

D

D

0

s

(g

s

2

C

C -

3

+

+

y

g

l

l

8 -

2

8

r

,

8

-

a

a

A

a

c

t

t

5

d

D

D

s

p

o

o

1

t

t

B

C

0

C

/

l

/

l

9

H

a

a

t

+

t

+

-

8

o

8

o

.

t

t

g

-

0

A

D

c

D

/ /

0

A

4

r

o

3

- C

2

C

e

-

s

r

c

+

4

+

,

p

e

t

p

8

B

8

+

+

9

y

s

1

t

1

D

H

n

D

9

o

9

C

C

e

1

n

1

i

-

-

e

+

t

+

3

3

0

g

7

7

0

a

8

8

5

2 2

2

3

2

1

-

1

- V

6 )

4

-

P

6

+ +

8

y

8

B

2

v

v

L

a

1

/

1

1

n

n

H

d

<3

d

U

e

c

e

c

,

o

(I

n

B

B

B

B

o

l

a

4 H

H

H

i

H

T

B 5

r

0

s

L 0

1

l 5

e

1 a

%

%

% %

5

A 3

y

b

x

-

c

+ +

ra ,

0

a

7

u

e 0

m .

1

d

a

h 2

u

9

c

r p

n

B

,

e

ri l

l

y

A

v

A

1

0

0

0

e 0

i e

0

1

0

0

1

N

N

P

L

c

0

1 0

5

g

g F

1

0 D

,

1

B A

A

e

e

s

V l

A

a

B

B

B N

F

H

H

H M

L

L

D

D

) l

T

T )

5

5

5

l

,

,

,

5

5 5

.

,

, ,

2

2 1

1

0

0 m

/

0

0

1

1

2

2 3

t

c m

.

/

s q

o

t

c q

r

o

e

s

o e

e

r

p o

(g

s

5

e p

- (g

s

s 7

5 -

g

y s

7

3

- 4

6

- r

a

g ,

A

y

- -

2

-

8

r c

1

d

↑ 5

s

,

a A

c

p

1

8

s d

B

0

p

D

2 B

9

H

0

N

H

9

e

n

A

p

o

n

i

y

e

2

t

o

i

5

p

rs

o

-

t

y -

e

rs

n t

g

t

0

5 g

v e

n

-

e o

A

0

8

n

7 -

5

n

v

A

g

r

- n

s

- -

e

2

2

8

- r

3

o

n

s c

i

e

,

, e

B

c i

c V

t

o

p

B

8

6

g

t

p

H

o

c

B

1

a

r

H

o

a

V

H

e

r

P

B

s

,

e

P

s

n

H

a

,

i

n

s

5

3

a

3

a 5

i

9

r

r

0

0

c

s 5

0 0

2

9

5

e

e

1

1

u

,

a 9

2

- -

8 +

y

+

,

y

x

b

x

b

a

c

0

6

5

a

8

a

7

9

c

u

5

m

.

.

m

d

1

d

,

a

u

2

u

7

y

c

B

n

n

B

8

,

l

l

A

l

l

A

4

y

A

A

e

e

N

,

8

N

1

0

0

0 0

0 N

1

0 N

0

0

c

c

F

e

0

1

0

3

0

1

F g

0 g

0 0

g

l

g

D

D

0

0 1

1

0 ,

0

a

B

B

A

A

A

0

1 A

1

0

e

1

e

V

s

e

V

s

l

A A

1

m

a

B

B

B

B

B

B

e

N

N

F

H

H

H

F

H

F

M

H H

L

L

D

D )

T

T

l

5

5

5

5

5

5

,

,

,

,

,

,

m

0

0

1

1

2

2

0

0

1

1

2

2

/

)

l

q

n

e

m

o

/

i

g

n

(

-

q

o

rs

5

i

g

e

e

2

s

7

-

A

-

r v

2

(g

-

6

8

5

s

,

e n

1

g

4

B

v

7

o

2

<3

A 7

2

n

c

H

,

-

6

s

-

2

o

o

4

r

B

c

A

1

<3

e

H

D

e

ro

s

p

e

e

y

s

p

t

4

1

y

o

5

t

0

t

n

t

2

o

2

3

2

e

1

n y

7

n

-

n

6

,

+ +

g

e a

2

6

e

e

g

d

i

i

V

<3

1

t

t

B

V

a

a

B

H

,

H

P

P

3

5

n

,

0

9

1

0

a

n

i

1

7

5

r

a

7

s

,

x

y

-

i

1

+ +

e

0

r

0

a

0

a

3

s 0

1

. b

8

7

c

e

1

7

d

a

3 -

3

+

y

7

u

x

b

m

c

,

9

a

1

a

u

u

2

m

.

2

d

c

n

a

2

u

1

B

,

l

c

B

n

l

A

y

A

,

l

e

A

l

6

N

y

A

N

c

e

2

N

g

5

g

F

1

0

0

N

D

0

0

c

,

0

0

0

0

1

F

3

B

0

0

A

g

0

A

1

g

1

0

0

0

D

e

1

0

0

,

e

s

V

l

A

1

0

0

B

A

A

1

0

e

1

0

a

B

B

B

e

s

V

l

1

A

N

1

a

B

B

B

F

H

H

H

M

N

F

H

H

H M

A L T

( I U/ L )

1A

64 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

2.34% 1.11% 0.20%

Liver e P

. r t e t

7 2 - 8

1 0.80% 0.55% 0.02% c B

H Peripheral Blood

CD8 Fitc 1B Day 0 Day 30 HBsAg-seroconversion C P A

1 9 1 - 3

8 Figure1.Patientcharacteristicsandexvivo 1 v

n frequenciesofHBVspecificCD8+cells. e B

H A)Biochemicalandvirologicalcharacteristics ofthepatients,andexvivofrequenciesof e P

7 HBVspecificCD8+cellsintheliverandPB, 2 - 8

1 obtainedinthestudypopulationatdif ferent c B timepointsduringacuteHBVinfection. H Frequenciesofcirculatingandintrahepatic CD8 Fitc HLA-A2 negative HLA-A2 positive HBc1827specifi c andHBenv183191 1C HBV patient non-HBV patient specificCD8+cellswereobtainedatfi rst presentation(day0),30daysafterfi rst presentation(patients1and3),atthetimeofHBsAgseroconversionand90dayspost HBsAgseroconversion(patients2,3and5).Thefrequencieswerecalculatedaspercentage HBcorHBenvtetramer+CD8+cellsofthetotalpopulationCD8+cellsintheliverandPB.The frequenciesofHBpol816824specificCD8+cellswerealwaysbelowthelevelofbackground stainingandarenotshowninthefigures.The“FNABcellnumber”indicatesthecellnumbers thatwereobtainedbyFNABateachtimepointandthatwereanalysedbyflowcytometry. ThePCRdetectionlimitwas362geq/ml.Thearrows(↑)indicatethetimepointsofsampling oftheliverandPBintheindividualpatients.ND=NotDetermined,TDL=TetramerDetection Limit;0.03%oftotalCD8+cells. B)FrequenciesofintrahepaticandcirculatingHBV sepcificCD8+cellsobtainedina representativepatient(patient1). Thenumberintheupperrightquadrantindicatesthe frequencyHBc1827specificCD8+cellsmeasuredatthecorrespondingtimepointdepicted atthebottomofthefigure. C)Resultsobtainedinrepresentativecontrolpatients.Theupperhalfofthefigurerepresents thebackgroundstainingfortheHBenv183191tetramerobservedinliverderived lymphocytesfromarepresentativeHLAA2negativepatientwithchronicHBVinfection,anda representativeHLAA2positiveliverdiseasepatientwithnoHBVinfection.Thebottomhalfof thefigureshowsthesamefortheHBc1827tetramer.

65 CHAPTER5 and30.1±1.0,respectively).Atalltimepoints,theratioofCD8+/CD4+cellsinthe liverwashigherthaninPB,indicatingcompartmentalizationofCD8+ Tcellsinthis organ(fig2a).Inthreepatients,thepeakratioofCD8+/CD4+cellswasobservedat fir stpresentation,andwhenpatientsweredividedonthebasisofserumALT atthis timepoint,therewasacorrelationbetweendegreeofliverdamageandtheratioof intrahepaticCD8+/CD4+cells(r=1.0,p<0.01)(fi g2b).Thesedatasuggestthat CD8+Tcellsareinvolvedindiseasepathogenesisthroughcytolyticliverdamage. s l l

e Liver Peripheral Blood c 6 6 + 4 5 5 D C

/ 4 4

+

8 3 3 D

C 2 2

o i

t 1 1 a

R 0 0 day 0 HBsAg-seroconversion day 0 HBsAg-seroconversion A Time point of sampling

3500 9 s l r = 1.0, p < 0.01 l 3000 8 e c 7 + 4

) 2500 L

6 D / C U

2000 I

5 / Ratio CD8+ / CD4+ cells in the liver (

+ T 1500 4 8

L Serum ALT (IU/L) at day 0 D A 3

1000 C

o

2 i t

500 a 1 R 0 0 Patient 1 3 5 2 High Low level of serum ALT at first presentation B Figure2.RatioofCD8+/CD4+cellsintheliverandPB. A) RatioofCD8+/CD4+cellsintheliverandPBatfi rstpresentation(day0)and atthetimeofHBsAgseroconversion. TheratioofCD8+/CD4+cellswasnot determinedat90daysafterHBsAgseroconversion. B) RatioofCD8+/CD4+cellsintheliverinrelationtoserum ALTlevelatfirst presentation.Onthehorizontalaxisinthegraphthepatientsarestratifi edon thebasisofserum ALTatday0,withdecreasingvaluefromlefttoright.On theverticalaxesthecorrespondingratioofCD8+/CD4+cellsintheliverat day0isdepicted,andtheserum ALTlevelsmeasuredatthistimepoint(paired Spearman’scorrelationtestr=1,0(p<0.01).Inpatient4theratioofCD8+/ CD4+cellswasnotdeterminedatfirstpresentationandthedataarethereforenot shown.

66 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

LongitudinalanalysisofthefrequencyofintrahepaticHBVspecificCD8cells To analyzethedynamicsofintrahepaticHBV specificCD8+ Tc ellresponses,at severaltimepointsduringacuteHBVinfectionliverderivedlymphocytesandPBMC fromthepatientsweredoublestaineddirectly exvivo,withantiCD8MAbsandwith HLAA2/peptidetetramericcomplexesabletovisualizeCD8+Tcellsspecificforcore 1827 (HBc 1827),envelope 183191 (HBenv 183191)andpolymerase8 16824 (HBpol816824).Figure1ashowsthefre quenciesofHBVspecificCD8+Tcellsin theliverandPBatthetimepointsofsampling.Figure1bshowsrepresentativeFACS results.Duringthecourseofinfection,intheliveroffourpatients (patient 1, 35) HBc1827+CD8+Tcellsweredetected,andinthreepatients (patient24)HBenv 183191+CD8+ Tc ellsweredetected .InnoneofthepatientsHBpol8 16824+ CD8+Tcellsweredetectableatanytimeduringinfection,eitherintheliverorPB . LongitudinalanalysisofthefrequencyofHBVspecificCD8+Tcellsduringrecovery fromtheinfection,asshownbyprogressivereductionandnormalizationofserumALT andHBsAgseroconversion,showedconsiderablevariationbetweentheintrahepatic compartmentandPB .Atalltimepointsduringinfectiontherewasse questeringof HBVspecificCD8+Tcellsintheintrahepaticcompartment,witha 1.2to21.7fold concentrationofHBc 1827orHBenv 183191specificcellsinthelivercompared toPB.ThepeakfrequencyofcirculatingHBVspecificCD8+Tcellsoccurredatfirst presentationanddeclinedincorrelationwithbiochemicalandvirologicalparameters. BythetimethatHBsAgwasundetectableintheserum,inthreepatients (patients1, 2and4)thepercentageofcirculatingHBc1827andHBenv183191specificCD8+ Tcellsdidnotexceedthebackgroundlevelof0.03%,asobservedincontrols(fig 1c). Incontrast,atthistimepointhighfrequenciesoftetramerpositivecells(0.32–1.86% oftotalCD8+Tcells)werestilldetectableintheliverofallpatients.Interestingly,inall patientssampled90dayspostHBsAgseroconversionthefre quencyofintrahepatic HBenv183191+CD8+TcellsandHBc1827+CD8+Tcellsremainedhigh (0.11 2.65%),suggestinganimportantroleofintrahepaticvirusspecificcellsatthisstage ofthedisease. TheabsenceofdetectableintrahepaticHBpol8 16824specificCD8+cellsmaybe causedbyviralmutationsresultinginspecificepitopeinactivation3,17.Therefore,inall fiv epatientsthisepitopewasse quenced.ThreepatientswereinfectedwithanHBV straincarryinganasparticacidtovalinemutationatposition820(patient2,3and4) whichmaybeassociatedwithreducedCD8+Tcellactivation.

HLADRexpressiononintrahepaticHBV specificTcellsduringresolutionofacute HBVinfection Figure3showsthepercentageofHLADRe xpressingHBVspecificCD8+Tcellsin theliverandPB.DuringtheacutephaseofHBVinfection,HBenv183191andHBc 1827specificCD8+ Tc ellshadanactivatedphenotypebecausealargefraction ofthesecellse xpressedtheactivationmar kerHLADR.Atthisstageofdisease,in allpatientsthepercentageofHLADRe xpressingHBVspecificCD8+ Tc ellswas higherintheliverthaninPB,suggestingtheirincreasedreactivityatthesiteofvirus

67 CHAPTER5

replication.Duringrecoveryfromtheinfection,withprogressivereductionofserum ALTlevels,expressionofHLADRintheliverandPBwasdownregulated.However, atthreemonthspostHBsAgseroconversionHLADRexpressiononHBc1827 specificcellsintheliverofpatient5hadexceededthelevelobservedduringthe acutephaseofinfection(24%vs.10%,respectively),inparallelwiththeincrementin HBc1827tetramerbindingalsoobservedinthispatient,suggestingreactivationof thesevirusspecificcells.

g g n i n i s s s s l l s e r e s e

l 70 r l c 40 p

p e x + x c e 60

35 8 e

+ R D

8 30 50 R D C

- D D

- 25 +

A 40 C r

A t L

+ 20 L e H r

t 30

t H

e

e 15 v t e g

n 20 g c a

10 e t a B B t 10 5 n n H e H e c 0 0 r c r 1 2 3 e 1 2 3 e P day 0 HBsAg- day 90 post

P day 0 HBsAg- day 90 post seroc. HBsAg-seroc. seroc. HBsAg-seroc.

Time point of sampling

Liver Peripheral Blood

Figure3.HLADRexpressiononHBVspecificCD8+cells. TheleftfigureshowsthepercentageofHLADRexpressingHBc1827specificCD8+ cellsmeasuredintheliverandPBofthestudypopulation(mean+SEM). Theright fig ureshowsthesamefortheHBenv183191specifi cCD8+cells.At90dayspost HBsAgseroconversiononlypatients2,3and5weresampled.

Discussion

Althoughseveralstudieshavepreviouslyshownimportantdifferencesbetween circulatingandintrahepaticHBV specificCD8+ TcellsinchronicHBVinfection 7 8, thisisthefi rstreportthatdescribesdifferencesinthecharacteristicsofthevirus specificCD8+ Tc ellresponseintheliverandPBduringhumanselflimitingacute HBVinfection. Interestingly,ourresultsshowacorrelationbetweenthedistributionofCD8+ Tcells intheliverandthedegreeofliverdamageatfi rstpresentation.Atthetimeofliver inflammationonlyafractionoftheintrahepaticCD8+ Tc ellsdisplayedtetramer binding,andthereforeourdatamay subscribetherelevanceofnonspecifi c CD8+ Tc ellrecruitmentinliverdamageinacuteHBVinfection.Moreover,animportant fin dingofthisstudyisthatwedidnotobserveaclearcorrelationbetweenthedegree

68 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

ofliverdamageandtheprofi leofexvivointrahepaticHBVspecificCD8+Tcells. Based upon studiesinanimalmodel s 1820andthefi ndingthatinthepre clinical incubationphaseofacuteHBVinfection,thereisalreadyalargefractionofcirculating HBVspecificTcellsprecedingma ximalliverdamage ,ithas been suggestedthat HBVspecificTcellsmayinfil tratetheliver wherethe y can contributetoviral suppression,andsubsequentlytriggeracascadeofeventsleadingtoliverdamage 21.Thiscascademayincluderecruitmentofa ctivatedCD8+Tcells,notaccounted forbytetramerstaining.However,alimitationofthetetramerte chnologyisthatit canonlybeappliedtostudydefin edepitopes.Althoughtheepitopesweusedinthis studyhavebeenchosenbecausetheyareamongthemo stfrequentCD8+Tcell epitopesfoundinHBVinfe ctionafterscreeningwithmultiplepeptides3,5,22,23,ithas alreadybeendescribedthatina cuteHBVinfe ctionthe Tcellresponseisdirected tom ultipleepitope s,includingtho senot covered byourtetramer s.Therefore, we may underestimatetheHBV specificCD8+Tcellre sponse 3, 4, 21.Amoredetailed longitudinalanalysisoftheintrahepaticevents,whichincludestheincubationphase andawidespectrumofTcellepitopes,isneededtofurtherclarifythemechanismof liverdamageinacuteHBVinfection. InallpatientshighfrequenciesofintrahepaticHBVspecificcellsremaineddetectable afternormalizationofserumALTwhereasthefrequenciesofcirculatingHBVspecific cellsdeclinedinparallel with serumALT.Sinceatalltimepoint sof samplingthe frequencyofHBVspecificCD8+Tcells washigherintheliverthaninPB ,their disappearancefromthe circulationma yindeed bee xplained by sequesteringat thesiteofviralrepli cation.Similarenrichmentofvirusspecificcellsintheliver,has previouslybeenfoundinpatientswithchronicHBVinfection7.Although,ithasbeen shownrecentlythatinnonhepatotropicvirusesactivatedprimaryormemoryCD8+ Tcellsmigratetotheliver 24,andthatinthenormalh umanliverC D8+Tcellsare presentatfre quencies whichareenri ched comparedtotho seinthe blood 25,our fin dingmaysuggestthatHBV specificCD8+Tcellscontributeto clearanceofthe virus. Ourdatadonote stablish whetherHBVspecificCD8+Tcells controlvir us replicationthro ughthe secretionof cytokinesalone,orin combination withdire ct lysisofinfectedcells.However,othergroupshavealreadyshownthatCD8+cellsare capableoftheseHBVspecificfunctionalresponses2628. Itha spreviously been shownthatina cuteHBVinfe ctionvir usspecificCD8+T cellsarestillpresentinthecirculationforseveralmonthsaftercompleteclinicaland serologicalrecovery3,26.Hereweshowthatthesecellscanalsobedetectedinthe liveratthreemonth spostHBsAgseroconversion,butathigherfre quenciesthanin PB.Interestingly,atthistimepointinonepatient boththefrequencyofintrahepatic HBc1827specificCD8+ cellsas wella sthelevelofHLA DRe xpression was increased, suggestingrea ctivationofvir us specific,presumablymemoryTcells. AC D8+Tcellresponsemaybemaintained bycontinuedantigenicstimulationby virusthathasnotyetbeeneradicatedfromtheliver .Thisconceptissupportedby thefindinginaprevio usstudy,thatinama jorityofpatientswithlonglastingTcell responsesinperipheralblood,minuteamountsofHBVDNAcouldbedetectedyears

69 CHAPTER5 afterseroconversion29. Inconclusion,ourmeasurementsofcirculatingandintrahepaticHBV specificCD8+ cellsinpatientswithselflimitingacuteHBVinfectionhavedemonstratedthatvirus specificcellsarecompartmentalizedintheliver .Theirsequesteringinthelivermay suggestaroleinresolutionoftheacuteinfection.Forseveralmonthsaftercomplete HBsAgseroconversionintrahepaticHBVspecificCD8+Tcellsremaindetectableat highfrequencies. Acknowledgements

Theauthorswishtothan kRehkaBindaandRaymondPotfortheirassistancein obtainingandprocessingtheFNABsamples.

70 CHAPTER5CHAPTER5INTRAHEPATICTCELLSINACUTEHBV

References 1. GuidottiLG,ChisariFV.Tokillortocure:optionsinhostdefenseagainstviralinfection.CurrOpinImmunol 1996;8:47883. 2. NayersinaR,FowlerP ,GuilhotS,etal.HLA A2restrictedcytotoxic Tlymphocyteresponsestomultiple hepatitisBsurfaceantigenepitopesduringhepatitisBvirusinfection.JImmunol1993;150:465971. 3. RehermannB,FowlerP,SidneyJ,etal.ThecytotoxicTlymphocyteresponsetomultiplehepatitisBvirus polymeraseepitopesduringandafteracuteviralhepatitis.JExpMed1995;181:104758. 4. MainiMK,BoniC,OggGS,etal.DirectexvivoanalysisofhepatitisBvirusspecifi c CD8(+)T cells associatedwiththecontrolofinfection.Gastroenterology1999;117:138696. 5. BertolettiA,CostanzoA,ChisariFV,etal.CytotoxicTlymphocyteresponsetoawildtypehepatitisBvirus epitopeinpatientschronicallyinfectedbyvariantvirusescarryingsubstitutionswithintheepitope.JExp Med1994;180:93343. 6. RehermannB,ChangKM,McHutchinsonJ,etal.DifferentialcytotoxicTlymphocyteresponsivenesstothe hepatitisBandCvirusesinchronicallyinfectedpatients.JVirol1996;70:7092102. 7. MainiMK,BoniC,LeeCK,etal.Theroleofvirusspecifi cCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 8. BertolettiA,D’EliosMM,BoniC,etal.DifferentcytokineprofilesofintraphepaticTcellsinchronichepatitis BandhepatitisCvirusinfections.Gastroenterology1997;112:1939. 9. BravoAA,ShethSG,ChopraS.Liverbiopsy.NEnglJMed2001;344:495500. 10. SprengersD,vanderMolenRG,KustersJG,JanssenHL.Coarsevs.fi neneedleaspirationbiopsy .J Hepatol2004;41:5034;authorreply504. 11. LautenschlagerI,HockerstedtK,HayryP.Fineneedleaspirationbiopsyinthemonitoringofliverallografts. TransplInt1991;4:5461. 12. SprengersD,vanderMolenRG,KustersJG,etal.Flowcytometryoffi neneedleaspirationbiopsies:a newmethodtomonitorintrahepaticimmunologicalenvironmentinchronicviralhepatitis.JViralHepat 2005;12:50712. 13. PasSD,FriesE,DeManRA,OsterhausAD,NiestersHG.Developmentofaquantitativerealtime detectionassayforhepatitisBvirusDNA andcomparisonwithtwocommercialassays.JClinMicrobiol 2000;38:2897901. 14. NiestersHG.Clinicalvirologyinrealtime.JClinVirol2002;25Suppl3:S312. 15. StuyverL,DeGendtS,VanGeytC,etal. An ewgenotypeofhepatitisBvirus:completegenomeand phylogeneticrelatedness.JGenVirol2000;81:6774. 16. NorderH,Courouce AM,MagniusLO.Completegenomes,phylogeneticrelatedness,andstructural proteinsofsixstrainsofthehepatitisBvirus,fourofwhichrepresenttwonewgenotypes.V irology1994; 198:489503. 17. MainiMK,ReignatS,BoniC,etal.TcellreceptorusageofvirusspecificCD8cellsandrecognitionofviral mutationsduringacuteandpersistenthepatitisBvirusinfection.EurJImmunol2000;30:306778. 18. MoriyamaT,GuilhotS,KlopchinK,etal.Immunobiologyandpathogenesisofhepatocellularinjuryin hepatitisBvirustransgenicmice.Science1990;248:3614. 19. NakamuraI,NuppJT,CowlenM,etal.Pathogenesisofexperimentalneonatalwoodchuckhepatitisvirus infection:chronicityasanoutcomeofinfectionisassociatedwithadiminishedacutehepatitisthatis temporallydeficientfortheexpressionofinterferongammaandtumornecrosisfactoralphamessenger RNAs.Hepatology2001;33:43947. 20. AndoK,Moriyama T, GuidottiLG,etal.MechanismsofclassIrestrictedimmunopathology .Atransgenic mousemodeloffulminanthepatitis.JExpMed1993;178:154154. 21. WebsterGJ,ReignatS,MainiMK,etal.IncubationphaseofacutehepatitisBinman:dynamicofcellular immunemechanisms.Hepatology2000;32:111724. 22. RehermannB,FerrariC,PasquinelliC,ChisariFV.ThehepatitisBviruspersistsfordecadesafterpatients’ recoveryfromacuteviralhepatitisdespiteactivemaintenanceofacytotoxic Tlymphocyteresponse.Nat Med1996;2:11048. 23. PennaA,ChisariFV,BertolettiA,etal.Cytotoxic TlymphocytesrecognizeanHLAA2restrictedepitope withinthehepatitisBvirusnucleocapsidantigen.JExpMed1991;174:156570. 24. MasopustD,VezysV ,UsherwoodEJ,etal. ActivatedprimaryandmemoryCD8 Tc ellsmigrateto nonlymphoidtissuesregardlessofsiteofactivationortissueoforigin.JImmunol2004;172:487582. 25. WardSM,JonssonJR,SierroS,etal.V irusspecificCD8+Tlymphocyteswithinthenormalhumanliver . EurJImmunol2004;34:152631. 26. UrbaniS,BoniC,MissaleG,etal.V irusspecificCD8+lymphocytessharethesameef fectormemory phenotypebutexhibitfunctionaldifferencesinacutehepatitisBandC.JVirol2002;76:1242334. 27. ThimmeR,WielandS,SteigerC,etal.CD8(+) Tcellsmediateviralclearanceanddiseasepathogenesis

71 CHAPTER5

duringacutehepatitisBvirusinfection.JVirol2003;77:6876. 28. MenneS,RonekerCA,RoggendorfM,GerinJL,CotePJ, TennantBC.Deficienciesintheacutephase cellmediatedimmuneresponsetoviralantigensareassociatedwithdevelopmentofchronicwoodchuck hepatitisvirusinfectionfollowingneonatalinoculation.JVirol2002;76:176980. 29. PennaA,Artini M,CavalliA,etal.Longlastingmemory Tc ellresponsesfollowingselflimitedacute hepatitisB.JClinInvest1996;98:118594.

72 CHAPTER 6

In vivo immunization following virus suppression: a novel approach for inducing immune control in chronic hepatitis B

D. Sprengers1, H.L.A.Janssen1, J.Kwekkeboom1, H.G.M.Niesters2, R.A. de Man1, S.W.Schalm1

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

JViralHepat2003:10:79 CHAPTER6

Despitethepresenceofaneffectivevaccinesince1982,HepatitisBstillranks amongthehighestcausesofmortalityfrominfectiousdiseasesworldwide. Thereis growingevidencethatanactiveantiviraltreatmentstrategyinthosewithprogressive diseaseassociatedwithactiveviralreplicationmayleadtosignifi cantreductionin morbidityandmortality1.OneapproachsimilartothestrategysosuccessfulinHIVis tosuppressviralreplicationwith(acombinationof)nucleosideanalogues;according tocurrentexperiencesuchtherapyneedstobegivenindefi nitely.Anotherstrategy aimsattheinductionofimmunecontrolbyatreatmentcourseoflimitedduration; thisapproach,ifequallyef fective,istobepreferredforobviousreasonssuchas compliance,sideeffectsandcosts. Thetwocurrentlylicenseddrugs:lamivudineandinterferonalphaareeachexamples ofthetwodifferentapproaches. Theireffectivenessislimitedeitherbythehigh incidenceofresistance(lamivudine)orlowincidenceofinducingimmunecontrol (interferon).Combinationofastandardcourseofinterferontherapywithlamivudine mayincreasetherateofinducingimmunecontrol 2,3,butasustainedtherapeutic response(HBeAgnegativity,HBVDNA<105geq/mlandALTnormality612months afterstoppingtreatment)isobservedinonlyaminorityofpatients. We observedthatcombininglamivudinewithinterferontherapyinducesamore rapiddeclineoftheviralloadthanmonotherapy 4.Weassumethatprofoundvirus suppressionmayallowrecoveryofthenonresponsivenessoftheeffectorarmofthe immuneresponse5;however,atthesametimeitmayweakentheantigenexpression oninfectedcellsthatneedtoberemovedbytheimmuneresponse. Invivoimmunization followingvirussuppressionaimstooptimi zeconditionsfor aneffectiveimmuneresponse:followingrapidandprofoundvirussuppressionby interferonlamivudinecombinationtherapy,lamivudineiswithdrawnintermittentlyfor 4weeksduringcontinuedinterferontherapy.Withdrawaloflamivudinewillleadto increasedviralreplicationininfectedcellswithstrongerantigenexpression,andtoa rapidriseinviralloadinthebloodwithsubsequentimmunestimulation.Weassume thatthecontinuationofinterferontherapywillhelpthedevelopmentofanimmune response,butmostimportantlywillpreventreinfectionofhepatocytesduring renewedactiveviralreplication. WehavetreatedtwochronichepatitisBpatients(♂21years,♂36years,bothHBeAg negativeandHBVDNApositive)withinvivoimmuni zationfollowingsuppressionof viralreplicationwithinterferonalphaandlamivudine.After612monthsofcombination therapylamivudinetherapywasinterruptedinitiallyfor2weeksandsubsequentlyfor 46weekswhileinterferontherapywascontinued.Inbothpatientswithprofound virussuppression(HBVDNA <103copiesperml)weobservedarapidreboundof viralreplicationuponwithdrawaloflamivudine;thepeakHBVDNA wassmallerafter thesecondperiodofwithdrawal.Risesinhepatitisactivityfollowedeachepisode ofrenewedviralreplication.AfterreintroductionoflamivudineHBVDNA became undetectablebyPCRfollowedbynormalizationofserumALT(figure1).

74 INVIVOIMMUNIZATION

IFN (30 MU/wk) IFN (30 MU/wk)

Lam (150mg/d) Lam (150mg/d) HBV DNA pcr + + + - HBV DNA pcr + + + - - 7 50 4 l 10 10 70 HBV DNA

m ALT /

q 106 e HBV DNA A g

40 ALT 60 L A T

N 5

10 I U D

/ L V 30 B 4 50

H 10

3 3 10 20 10 40 0 8 16 24 32 40 48 0 16 32 48 64 80 88

Weeks after start of therapy A B Fig.1. Assessmentofviralload(HBVDNA)andhepatitisactivity(ALT)intwopatientswith chronicHBVinfectionun dergoinginvivoimmunization followingviralsuppression . HBVDNAwasquantifiedusingHBVmonitorassa y(RocheDiagnostics;detection limit1x103geq/ml).Below103geq/mlHBVDNAwasmeasuredbyankineticPCR (TaqManbase dassay; detectionlimit 373geq/ml). BaselineH BV DNA:A: ♂ 21y 9,9x106geq/ml,B:♂36y1,6x105geq/ml. PatiëntAshowsacharacteristicbiphasicdeclineofHBVDNA,andmarkedrebound uponstoppinglamivu dine.Thesecon drebound waslo werthanthe firstan d associatedwithsomehepatitisactivity. PatiëntB,withaHBVDNAabove105geq/mlatstart,showedarapiddeclineandHBV DNAwasundetectablebytheRocheassayforayear.UponstoppinglamivudineHBV DNAreboundedtwice;thesecondpeakwaslowerthanthefirst.Hepatitisactivitywas associatedwithbothflares.FinallyHBVDNAbecamenegativebyPCR.

Theseobservationsareinouropinionastrongstimulustofurtherexploretheconcept ofinvivoimmunization.Manyfactorsneedtobeoptimizedlikethedurationofvirus suppressionpriortowithdrawal, thedurationoflamivu dinewithdrawal,thenumber of withdrawals,the doseofinterferonrequired topreventinfectionofhepatoc ytes withoutinterferingwiththeimmuneresponse. MonitoringofHBVspeci fic CD8 cytotoxicandCD4helpercellsina dditiontosensitivemeasurementsofH BVDNA appearsessentialforansweringthesequestions. Conceptuallyinvivoimmunizationdiffersfromstructuredtreatmentinterruptions,as describedinHIVinfection6.Treatmentisnotinterruptedbutforonlyonecomponent oftherapy.Weassumethatthecontinuationofinterferontherapywillkeepuninfected hepatocytesintheirantiviralstate’andthatthepoolofinfectedhepatocytescontinues todecrease;incaseoftreatmentinterruptiononemustassumearapidspreadofthe

75 CHAPTER6

infectionandanincreaseinthenumberofinfectedcells. Invivoimmunization isalsodifferentfromcurrentl ydesignedtherapeuticvaccines. TheimmunesysteminpatientswithchronichepatitisBisonlyslightlyimpaired;the impairmentinimmuneres ponsivenessishe patitis Bspecific ratherthan general. Thereise videncethattheimmuneres ponsivenessactuall yishepatitis Bstrain specific,sincepatientswithchronichepatitisBapparentlycanclearasuperinfection withanotherhe patitisBstrain 7.Therapeuticvaccinesusuallyrestricttheanti genic profiletoafewepitopesthou ghttobeessential 8; presentlythereislimited proof abouttheireffectivenessingeneratingacellularimmuneresponse9.Sinceimmune responsesinresol vedhepatitisBinfectionsareoftendirecteda gainstseveralbut variableepitopes10,webelievethatthebestimmunestimulusisthewholeviruswith averybroadantigenicprofile.Furthermore,liveattenuatedvaccinespermitefficient MHCclassIpresentationofantigentostimulateCTL’s11.

Immunestimulationb ythecurrenta pproachof withdrawaloflami vudinedurin g continuedinterferonthera pymaysufficeinsome patients;itisconceivablethat inman yothersadditionalad juvantthera pyisneeded,forinstancetoshiftthe CD4responsetoa Th1profileortomaintaintheH BVspecific Thelperresponse ofsuf ficientduration. Webelie vethisconce ptofin vivoimmuni zationasano vel therapeutica pproachforchroniche patitis Bmeritstobein vestigatedfurtherina cooperativeeffortbetweenacademicinstitutionsandpharmaceuticalindustry.

76 INVIVOIMMUNIZATION

References 1. NiederauC,Heintges T, LangeS,GoldmannG,NiederauCM,MohrL,etal.LongtermfollowupofHBeAg positivepatientstreatedwithinterferonalfaforchronichepatitisB.NEnglJMed.1996,30;334:14227. 2. SchalmSW,HeathcoteJ,CianciaraJ,FarrellG,ShermanM,WillemsB,etal.Lamivudineandalphainterferon combinationtreatmentofpatientswithchronichepatitisBinfection:arandomisedtrial.Gut.2000;46:5628. 3. BarbaroG,ZechiniF,Pellicelli AM,FrancavillaR,ScottoG,BaccaD,etal.Longtermeffi cacyofinterferon alpha2bandlamivudineincombinationcomparedtolamivudinemonotherapyinpatientswithchronichepatitis B.AnItalianmulticenter,randomizedtrial.JHepatol.2001;35:40611. 4. vanNunenAB,HansenBE,MutimerDJ,NiestersHG,deManRA,SchalmSW.Viralkineticsduring16weeks ofinterferonandlamivudinemonotherapyversusinterferonlamivudinecombinationtherapyinchronichepatitis Bpatients.Hepatology2000;32:379A. 5. BoniC,PennaA,OggG,BertolettiA,PilliM,CavalloCetal.Lamivudinetreatmentcanovercomecytotoxic TcellhyporesponsivenessinchronichepatitisB:Newperspectivesforimmunetherapy.Hepatology2001;33: 963971. 6. CarcelainG, TubianaR,Samri A,CalvezV,DelaugerreC, AgutHetal. TransientmobilizationofHuman immunodeficiencyV irus(HIV)specific CD4T helpercellsfailstocontrolvirusreboundduringintermittent antiretroviraltherapyinchronicHIVtype1infection.JVirol2001;75:234241. 7. KaoJH,ChenPJ,LaiMY ,ChenDS. AcuteexacerbationsofchronichepatitisBarerarelyassociatedwith superinfectionofhepatitisBvirus.Hepatology.2001;34:81723. 8. MichelML,PolS,BrechotC,TiollaisP.ImmunotherapyofchronichepatitisBbyantiHBVvaccine:frompresent tofuture.Vaccine2001;19:23952399. 9. HeathcoteJ,McHutchisonJ,LeeS,TongM,BennerK,MinukG,etal.ApilotstudyoftheCY1899Tcellvaccine insubjectschronicallyinfectedwithhepatitisBvirus. TheCY1899 TC ellVaccineStudyGroup.Hepatology . 1999;30:5316. 10. RehermannB,FowlerP,SidneyJ,PersonJ,RedekerA,BrownM,etal. ThecytotoxicTlymphocyteresponse tomultiplehepatitisBviruspolymeraseepitopesduringandafteracuteviralhepatitis.JExpMed.1995;181: 104758. 11. SederRA,HillAVS.Vaccinesagainstintracellularinfectionsrequiringcellularimmunity.Nature2000;406:793 798.

77

CHAPTER 7

In vivo immunization in combination with peg-interferon for chronic hepatitis B virus infection

D.Sprengers1,R.G.vanderMolen1,R.Binda1,J.G.Kusters1,R.A.deMan1,H.G.M. Niesters2,S.W.Schalm1,H.L.A.Janssen1.

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

Submitted CHAPTER7

Abstract

OnlyinaminorityofpatientswithchronicHBV(CHB)infectionwilltreatment withIFNαornucleosideanaloguesleadtosustainedvirologicalresponse. Invivo immunization (IVI)followingvirussuppressionaimstooptimi zeconditionsforan effectiveimmuneresponse :followingrapidandprofoundvirussuppression by interferonlamivudinecom binationtherapy ,lamivudineiswithdrawnintermittently duringcontinuedinterferontherapy.Itisthoughtthatwithdrawaloflamivudinewill leadtoincreasedviralreplicationandincreasedantigenexpressionwithsubsequent immunestimulation. TheaimofthisprospectivepilotstudywastoevaluateIVIasatherapeuticapproach forCHB.FourteenHBeAgpositiveCHBpatientsweretreatedfor 42weekswith acom binationofpegylatedinterferon alpha 2bandlamivudine. After 12weeks ofcom binationtherapylamivudinewaswithdrawnintermittentlyfor 3consecutive periodsof4weeksuntilitwaspermanentlystoppedinweek36. Attheendoffollow up(week52)allpatientshadremainedHBeAgpositiveandthe medianviralloadwassimilarto baseline.Duringtheinitial 12weeksoftreatment , therewasareductionof boththeHBV specific proliferationcapacityof Thcells andthefre quenciesIFNγproducingcells.Duringthelamivudineinterruption cycle therewasaninverserelation betweentheincreaseofHBV DNA,andthedecrease inproliferationcapacityandfre quencyofIFN γproducingcells.Theintrahepatic fractionofCD8+Tcellsincreasedduringlamivudinewithdrawal. Inconclusion ,IVIwasa bletotransientlystimulatetheHBV specific immune responsivenessof Tcells, butthemagnitudeoftheresponsewasinsuf ficientto causeabeneficialvirologicaleffect.

80 CHAPTER7CHAPTER7IVICLINICALSTUDY

Introduction

ChronichepatitisB(CHB)ischaracterizedbyinflammatoryliverdiseaseofvariable severityandisassociatedwithasignificantlyincreasedriskofdevelopingcirrhosis, liverfailureandhepatocellularcarcinoma1,2.Activeantiviraltreatmentofpatientswith progressivediseaseandactiveviralreplicationmayleadtosignifi cantreductionin morbidityandmortality 3.OneapproachsimilartothestrategysosuccessfulinHIV istosuppressviralreplicationwith(acombinationof)nucleosideanaloguessuchas lamivudine,adefovirdipivoxilorentecavir.Sincetheseagentsusuallydonotinduce immunecontrol,inmostpatientstheyneedtobegivenindefi nitely.However,lomg termeffectmaybelimitedbytheincidenceofresistance 4,5.Anotherstrategyaims attheinductionofimmunecontrolbyatreatmentcourseoflimitedduration. This approach,ofwhichinterferonalpha(IFNα)therapyisthegenerallyusedexample, istobepreferred.However,inonlyaminorityofpatientsconventionalorpegylated IFNα(PegIFNα)therapyleadstosustainedvirologicalresponse 6,7 .Although severalothercomponentshavebeeninvestigatedtoachieveimmunecontrolover thevirusinthepast 8,recentlymostattentionhasbeenfocusedonvaccinetherapy forCHB.TherapeuticvaccinationstrategiesaimatinducingorincreasingtheHBV specificCD8+andCD4+ Tcellresponsesthatareweak,antigenicallyrestrictedor undetectableinthebloodofchronicallyinfectedpatients.However,untilnowstandard antiHBVvaccinationaswellasDNA basedvaccineshavemostlynotbeenableto inducetheclearanceofthehepatitisBvirusinhumans 9,10.Therapeutic vaccines usuallyrestricttheantigenicprofi letoafewepitopesthoughttobeessential 11. SinceimmuneresponsesinresolvedhepatitisBinfectionsareoftendirectedagainst severalbutvariableepitopes 12,thebestimmunestimulusmaybethewholevirus withaverybroadantigenicprofile. Invivoimmunization(IVI)followingvirussuppressionaimstooptimizeconditionsfor aneffectiveimmuneresponse :followingrapidandprofoundvirussuppressionby interferonlamivudinecombinationtherapy,lamivudineiswithdrawnintermittentlyfor 3consecutiveperiodsof4weeksduringcontinuedinterferontherapy . Withdrawal oflamivudinewillleadtoincreasedviralreplicationininfectedcellswithexportof completevirusparticles,arapidriseinviralloadandincreasedantigenexpression onthehepatocytemembrane,withsubse quentimmunestimulation.Itisassumed thatthecontinuationofinterferontherapywillhelpthedevelopmentofanimmune response,andwillpreventreinfectionofhepatocytesduringrenewedactiveviral replication1315.IthasrecentlybeenreportedthatIVIwasabletotransientlysuppress viralreplicationintwopatientswithCHB 16.These observationswereastrong stimulustofurtherexploretheconceptof IVIasatherapeuticapproachforchronic HBVinfectioninaprospectivepilotstudy.

81 CHAPTER 7 IVI CLINICAL

Patients and Methods

Patients and study design Seventeen patients with CHB and alanine transaminase (ALT) levels greater than normal (>45 IU/l) were included in the study. All patients were HBsAg positive >6 months, HBV-DNA positive (>105 geq/ml), anti-HBV core antigen (HBcAg) antibody positive, and HBeAg positive. Patients had not received anti-HBV therapy within 6 months prior to enrollment. None of the patients had significant co-morbidity or cirrhosis. Antibodies to HCV, HIV-1, HIV–2 and hepatitis Delta were absent. Patients were treated for 42 weeks with a combination of 100μg pegylated interferon-alpha 2b (Peg-IFNα) weekly and 100mg lamivudine once daily. After 12 weeks of combination therapy lamivudine was withdrawn intermittently for 3 consecutive periods of 4 weeks until week 36, after which it was permanently stopped. Continuous Peg-IFNα therapy was given until week 42. There was a post-treatment follow-up period of 10 weeks, until week 52. Figure 1 shows the treatment schedule. Patients exhibiting HBeAg loss within the first 12 weeks were excluded from in vivo immunization and offered continuous PEG-IFNα and lamivudine therapy up to 12 months. The primary outcome measure was loss of HBeAg from serum. Secondary outcome measures were HBV- DNA levels below the level of detection in PCR assay, emergence of of mutations in the YMDD motif of HBV polymerase at week 42 and induction of an HBV-specific type 1 immune response. All patients signed informed consent before enrolment. This study was performed in agreement with the local Medical Ethics Committee.

��������� ��������������������������������������������������������������� ������������������������������������������������������

������� ����������� ���������

���������� �����������     ������������� ���� ������������� ���� ���� Fig 1. Study design. Patients were treated for 42 weeks with a combination of 100μg pegylated interferon- alpha 2b weekly and 100mg lamivudine daily. After 12 weeks of combination therapy lamivudine was withdrawn intermittently for 4 weeks until week 36, after which it was permanently stopped. Continuous Peg-IFNα therapy was given until week 42. The follow up period lasted until week 52. Virological assessment Blood samples for reported virological and biochemical data were obtained every 4 weeks. Serum HBeAg, HBsAg, anti-HBs, anti-HBc, anti-delta, anti-HCV and anti-HIV1/2 were determined using commercially available immunoassays (Abbott Laboratories, North Chicago, IL, USA). Serum HBV-DNA was measured with TaqMan

82 CHAPTER 7 IVI CLINICAL STUDY

real-time PCR (detection limit 100 geq/ml) validated on the Eurohep Standard 17. HBV-DNA was extracted from serum samples by using a MagnaPure LC station (Roche Applied Science, Penzberg, Germany). For sequencing, primers were used as described previously 18. The genotype of the predominant HBV population was determined by analysis of sequenced portions of the core and surface antigen encoding DNA isolated from the serum 19. At week 0 and 42 HBV polymerase mutant analysis was performed on HBV-DNA using a Line Probe assay (INNO-LiPA HBV DR; Innogenetics N.V., Gent, Belgium) 18.

Standard tissue needle biopsy and fine-needle aspiration biopsy of the liver Liver histology was assessed at baseline and at the end of Peg-IFNα treatment. The biopsies were scored centrally by an experienced pathologist who was unaware of the chronological order of the biopsy. Histological scoring was performed according to the histological activity index, as described by Ishak 20. Improvement of histology was defined as a reduction of at least two points for the necroinflammatory score (range 0-18) and one point for the fibrosis score (range 0-6). Optionally, a fine-needle aspiration biopsy (FNAB) was combined with a standard tissue biopsy at baseline and at the end of treatment (42 weeks), and additionally at 12 and 16 weeks (fig 1). Details of the FNAB-procedure are described elsewhere 21. Briefly, a mandarin containing 25- gauge (diameter 0.5 mm) needle (Braun AG, Melsungen, Germany) is punctured in the 8th or 9th right intercostal space. After removal of the mandarin a 10 mL syringe filled with 2 mL RPMI supplemented with 0.1% human serum albumin (Sanquin CLB, Amsterdam, the Netherlands) and 25 IU/mL heparin (Leo Pharma, Breda, the Netherlands) is attached. Liver cells are aspirated by negative syringe pressure. Next, the specimen is transferred into a 15 mL tube and placed on ice immediately before further analysis. Validation for immunological assessment of the liver using the fine-needle aspiration biopsy-aspirate, has been described previously 22.

Isolation of the PBMCs and flow cytometric Analysis At the start of therapy (0 weeks), at 12 weeks and at every 4 weeks thereafter until the end of the study (t=52wks) PBMCs from individual patients were obtained by ficoll separation (Ficoll-PaqueTM plus, Amersham Biosciences, Buckinghamshire, UK). The PBMCs were immediately frozen in medium containing 10% DMSO and stored at –135°C until further use. Flow cytometric analysis was performed on the stored samples and freshly obtained FNAB-cytology using fluorochrome conjugated antibodies specific for the surface markers CD4 and CD8 (Dako, Glostrup, Denmark). After staining the cells were analyzed using a four-color cytometer (FACScaliburTM, CELLQuest ProTM software, Beckton Dickinson Biosciences; San Jose, USA).

Proliferation assay The proliferative capacity of Th cells was determined as reported previously 23. Isolated PBMCs of all time points investigated were cultured in triplicate in a concentration of 1X105 cells per well in 100 μl RPMI 1640 (Bio Whittaker, Verviers,

83 CHAPTER 7 IVI CLINICAL

Belgium) containing 5% pooled human serum (Dept. of Immunohematology and Bloodbank, Leiden University Medical Center, Leiden, The Netherlands) and penicillin/ streptomycin (Gibco, Paisley, UK). The cells were stimulated with 1 μg/ml HBV core antigen (HBcAg) (Biomerieux, Boxtel, The Netherlands), 5 μg/ml Phytohemagglutin (Murex, Paris, France), or not stimulated and cultured for 6 days. After 5 days of incubation the cells were pulsed with 0.25 μCi/well of [3H]thymidine (Radiochemical Centre, Amersham, Little Chalfont, UK). The cells were harvested 16 hours later. Proliferation, was determined by liquid scintillation counting of the harvested cells and expressed as stimulation index (SI). The SI was calculated from the counts per minute found with antigen divided by that found without antigen.

Elispot assay Detection of IFNγ (U-CyTech, Utrecht, The Netherlands) was performed using an ELISPOT kit according to the manufacturer’s instructions. Of all time points investigated, cultures of PBMCs were established in triplicate on round bottom 96-wells plates (Corning Inc., New york, USA), in a concentration of 1X105 cells per well in 100 μl RPMI 1640 (Bio Whittaker) containing 5% pooled human serum (Leiden University Medical Center) and penicillin/streptomycin (Gibco). The cells were stimulated with 1 μg/ml HBV core antigen (HBcAg) (Biomerieux), 5 μg/ml Phytohemagglutin (Murex), or medium only (negative control) for 24 hours at 37 degrees Celsius, after which they were transferred into flat bottom, anti- IFNγ-coated 96-wells plates (Nunc A/S, Roskilde, Denmark) for another 24-hours incubation period. Cytokine spots were visualised by biotin-labelled antibodies and their numbers were established by using a Bioreader 3000 from BioSys (Karben, Germany).

Statistical analysis Flow cytometry data, cytokine profile and proliferation capacity were compared using the Wilcoxon matched pairs signed sum test. The paired Pearson correlation test was used to correlate reported data. For these analyses SPSS 11.5 for Windows (SPSS, Chicago, IL) was used. Where applicable data are reported as mean ± standard error of the mean (SEM).

Results

Patients and side effects Seventeen HBeAg positive patients were enrolled in the study (the intention to treat population). After 12 weeks of treatment with continuous lamivudine and Peg-IFNα combination therapy, 3 patients (18%) had lost HBeAg and these patients were excluded from IVI and offered continuous PEG-IFNα and lamivudine therapy up to 12 months. Fourteen patients (10 male) with a median age of 26 (22 – 61) were evaluated for the effect of IVI (the per protocol population) (table 1). The side effect profile in the 14 IVI patients and the 3 patients that received continuous PEG-IFNα and lamivudine therapy was similar, and it resembled data reported in previous clinical studies using

84 CHAPTER7CHAPTER7IVICLINICALSTUDY

PegIFNα7.Commonsideeffectsincludedflulikesymptoms,headache,fatigueand localreactionattheinjectionsite,andallwerereversible.

Table I. Per protocol population characteristics at inclusion.

Patient Sex Age ALT HBV DNA Genotype (years) (IU/L) (cp/ml) 1 M 47 136 4.7x108 D 2 M 30 48 5.1x109 D 3 F 31 74 1.4x109 B 4 F 22 301 7.8x108 B 5 M 23 82 9.8x108 C 6 M 26 56 3.4x1010 E 7 M 26 174 7.7x108 D 8 M 61 57 7.0x109 A 9 M 23 69 8.3x107 C 10 M 24 57 1.9x109 B 11 M 26 46 1.4 x1010 D 12 M 22 250 1.3 x1010 D 13 F 28 47 3.3 x106 D 14 F 26 46 1.7 x109 D median age: 26y (22 – 61) median HBV DNA: 1.3x109 cp/ml (3.3x106 - 3.4x1010) median ALT: 63 IU/l (46 – 301)

ALT, alanine aminotransferase.

Viralkineticsduringthestudy Attheendoffollowup(52wks)all14patientshadremainedHBeAgandHBVDNA positive,andtherewerethusnovirologicalresponders.Atbaselinethemedianviral loadwas1.34x10 9cp/ml(3.3x10 63.4x109)andat52weeksitwas2.0x10 8cp/ml (5.0x1041.1x10 10)(Fig2a). After12weeksofcontinuousPegIFNα–lamivudine

combinationtherapyHBVDNAhaddecreasedbyamedianof4.4log10(5.9–2.7)to 5.5x104cp/ml(1.0x10 3–1.2x106).Whenlamivudinewaswithdrawnduringthefi rst

interruptionof4weeks,medianHBVDNA levelsincreasedby3.3log 10(1.8–4.7) to1.3x108cp/ml(4.0x1033.5x109).Reintroductionoflamivudineat16wksreduced 5 3 7 theviralloadby2.7log 10(1.1–3.7)to2.4x10 cp/ml(1.0x10 –1.2x10 )within4 weeks.Asimilarpatternofresponsewasobservedduringandafterthesecondand thirdinterruptionoflamivudinetherapywithoutasignifi cantfurtherreductionofthe medianHBVDNAlevels.Permanentcessationoflamivudineatweek36resultedin anincreaseinmedianHBVDNA levelsfrom8.5x10 5(1.0x103–1.2x107)to4.8x10 7 3 10 cp/ml(1.0x10 –1.5x10 )atweek42,andafurtherriseof0.64log 10(3.8–3.0) occurredafterdiscontinuationofPegIFNαatweek42.In2patients(patient12and 14;14%)attheendofthestudytheviralloadhadstabilizedatasignificantlyreduced levelascomparedtobaseline(1.3x10 10and1.7x109vs.2.6x105and6.1x105cp/ml, respectively)(fig2b).

85 CHAPTER7

ALTresponse Attheendoftreatmentthemedian ALTlevelhadreducedfrom63IU/L (46–301) atbaselineto38IU/L (19436)at42wks(fi g 2a).There wasnoclearcorrelation betweenmedianALTandHBVDNAlevelsduringthecourseofthestudy(r=0.33, p=0.35).Theoretically,anHBVspecificimmuneresponseinducedbythetreatment regimen,mayresultinhepatitis.In2of14patients(patient12and14;14%)there wasaclearhepatitisflare(ALT≥100IU/L)during IVI.Inbothpatientsthehepatitis activityappearedtoberelatedtowithdrawinglamivudinetherapy,withtemporalALT risesandinpatient14transientviralsuppressionfollowingthetreatmentinterruptions (fig2b).In4patients(patient1,6,11and12;29%)areversiblehepatitisflareoccurred afterstoppingbothlamivudineandPegIFNαtherapy.

Peg-IFNPeg- γ

Lamivudine

10 1.0x10 80

1.0x108 /ml) 60 cp 1.0x106 40 1.0x104 ALT (IU/L)

2 20 HBV DNA ( 1.0x10

0 1.0x10 0 0 4 8 12 16 20 24 28 32 36 42 52

Week HBV DNA ALT

Peg-IFNPeg- γ

Lamivudine 400 IU/l 1.0x10 12 280

1.0x10 10 240

8 200 cp cp/ml) 1.0x10 160 1.0x10 6 120 ALT (IU/L) ALT (IU/L) 1.0x10 4 80 HBV DNA ( 1.0x10 2 40

0 1.0x10 0 0 4 8 12 16 20 24 28 32 36 42 52 Week Pt 12 HBV DNA Pt 14 HBV DNA Pt 12 ALT Pt 14 ALT Fig2.Assessmentofviralload(HBVDNA)andhepatitisactivity(ALT). 2a)MedianHBVDNAandALTlevelsofthepatientpopulationduringthestudy. 2b)HBVDNAandALTlevelsofpatients12and14.Inbothpatientstherewasmarked hepatitisactivityduring IVI,andinpatient14transientviralsuppressionduringthe secondlamivudineinterruption.Attheendofthestudy,inbothpatientstheviralload hadstabilizedatasignificantlyreducedlevelascomparedtobaseline.

86 CHAPTER7CHAPTER7IVICLINICALSTUDY

LongitudinalanalysisofHBVspecificThelpercellproliferationandIFNγproduction Ap roliferationassaywasperformedtomeasuretheimmuneresponsivenessofTh cellstoHBcAglongitudinallyduringIVI.Inthefirst12weeksoftreatment,therewasa progressivereductionofthemeanproliferationcapacityofThcellsfromaSIof5.7(± 2.5)to3.3(±0.9)(fig3a).Duringthefirstlamivudineinterruptioninallpatientsthere wasafurtherdecreaseinproliferationcapacitytoameanSIof 0.6(±0.2).When lamivudinewasreintroducedatweek16inallpatientstheHBVspecificproliferation capacitypartlyrestoredtoameanSIof3.5(±1.6)atweek20.Infact,alsoduringand afterthesecondandthirdlamivudineinterruptionanincreaseinproliferationcapacity coincidedwithdecreasingHBVDNA levelsandviceversa .Toanalyzethe kinetics ofantiviralTh1activitythefrequencyofIFNγproducingcellsafterinvitrostimulation withHBcAg,wasmeasuredlongitudinallyduringtherapy(fig3b).Inthefirst12weeks ofcontinuouslamivudine– PegIFNαcombinationtherapythemeanfre quencyof IFNγproducingcellsdecreasedinallpatients (85±36and 3±1.5IFNγproducing cells/1x105cells,respectively).Duringthelamivudineinterruptionsthecourseofthe frequencyofIFNγproducingcellswassimilartotheThcellproliferativeresponse (r =0.7,p=0.038);therewasaninverserelation betweenthelevelofHBVDNA and thefrequencyofIFNγproducingcellsinthesecondandthirdlamivudineinterruption. Therefore, IVIappearedtostimulatetheHBVspeci fic immuneresponsivenessof Thcells,butthemagnitudeoftheresponsewaslow ,andcomparedto baselineat theendoftherapyinnoneofthepatientsthevirusspeci ficproliferationcapacityof Thcells,ortheircapacitytoproduceIFNγhadimproved.

Peg-IFNα Peg-IFNα s

l Lamivudine Lamivudine l e

7 c 100 5 0 x

6 1 e x d 1

80 / n i 5 s l l n e o i c

t 4 60 g a l n i u

3 c u

m 40 i d t o

2 r S p

γ 20

1 N F I 0 0 0 4 8 12 16 20 24 28 32 36 42 52 0 4 8 12 16 20 24 28 32 36 42 52 Week Week

Fig 3.LogitudinalanalysisofHBV specific ThelpercellproliferationandI FNγ production 3a)Meanproliferationcapacity (±sem)ofThcellsobtainedduringthestudy,after stimulationwithHBcAg. 3b) Meanfre quency (±sem)ofIFNγproducingThcells duringthestudy ,after stimulationwithHBcAg.

87 CHAPTER7

Histologyandintrahepaticimmuneresponse Dataonthechangeinhistologyoftheliverbetweenbaselineandendoftherapy wereavailablefor13of14patients.Fibrosisscoresimprovedin1patient(8%)and worsenedin5patients(38%).Improvementsininflammatoryscoreswereseenin4 patients(31%)andinflammationhadincreasedin3patients(23%).In9patientsthe intrahepaticimmuneresponsewasmonitoredbyFNABatweek0and12,afterthe fir stlamivudineinterruption(week16)andattheendoftherapy(week42).Figure4 showsthatcomparedtoweek12,atweek16theratioCD8+/CD4+cellsintheliver hadincreasedby26%(1.5±0.08and1.9±0.1 1,respectively,p>0.05). Thisrise inthefractionofCD8+ TcellsoccurredinparallelwithincreasedHBVDNA levels andpresumablystrongerantigenexpressionbyhepatocytesduetolamivudine interruption.Interestingly,inpatient12and14therewasarelativelystrongriseofthe fractionofintrahepaticCD8+Tcells(2xthebaselinevalue;fig4).

4

3

2

1 Rato CD8+ / cells CD4+ CD8+ Rato

0 0 12 16 42

Week Study population Pt 12 Pt 14

Fig4.RatioCD8+/CD4+cellsinliverderivedlymphocytesobtainedatbaseline (week0),after12weeksofcontinuouslamivudine–PegIFNαtherapy(week12), afterthefirstlamivudineinterruption(week16),andattheendoftherapy(week42), inthestudypopulation(mean±sem)andinpatients12and14.

Lamivudineresistance Onepatient(patient11)hadpreviouslybeentreatedwithlamivudineandwasknown toharbora YMDDmutantthatwasalsoidentifi edafterIVI.All otherpatientswere treatmentnaïveanddidnotharbora YMDDmutantpriortoinclusion.Ofthisgroup, afterIVIinonepatient(patient3)aYMDDmutantwasdetected.

88 CHAPTER7CHAPTER7IVICLINICALSTUDY

Discussion

Althoughitwaspreviouslyreportedthat IVIwasa bletotra nsientlysuppressviral replicationintwopatientswithCH B16,inthecurrentpilotstudywewereu nableto showa beneficiale ffecto ntherespo nserate , comparedto continuousPeg IFNα –lamivudine combinationtherapyorPeg IFNαmonotherapy for 52weeks 7. Oneexplanationforthea bsenceofasustainedvirologicalresponseinthecurrent study,maybethati nmostpatientswewereu nabletorea chsufficientlylowlevels ofHBVDNA priortothela mivudinei nterruptions, forasig nificantH BVspecific immunerespo nsetodevelop .Inanimal modelshightiterso fvirala ntigenshave beenshowntonegativelyaffecttheantiviralimmuneresponse24.Previousstudies conductedinpatientswithchronicHBVinfectiontreatedwithIFNαmonotherapy25, lamivudine 26,oracombinationtherapyofIFNαandlamivudine 27,suggestthat T cellresponsivenessmaybeenhancedaftersignificantreductionsinviremialevelsto <105cp/ml.WhereasinthepreviousreportonIVI16bothpatientshadundetectable HBVDNAlevelsbeforelamivudineinterruption,inthecurrentstudyafter12weeks oflamivudinePegIFNαcombinationtherapyin9patientstheviralloadwasabove thisthreshold.Therefore,futureattemptstoachieveimmunecontroloverthevirusby IVImaybemoresuccessfulwhen,priortothenucleosideanalogueinterruptions,the HBVDNAlevelsaremoreprofoundlyreduced,andwhentheperiodsofinterruption areshortenedtolimittheviralrebound. Recently,ithasbeensuggestedthatchronicpatientswithhighviralloada ndthose whoharbortheH BVgenotypesCorDdisplaylowerrespo nseratestoPeg IFNα therapythanindividualswithHBVgenotypesAorB7.Inourstudy,themedianlevel ofHBVDNAwas1.3x109cp/mlandonly4of14patientsharboredanHBVstrain withgenotypeAorB.Althoughthe mechanismbywhichthege notypeaffectsthe immunomodulatorypropertieso fIFNαisyetunknown,boththehighviralloada nd thelargefractionofpatientswithgenotypesCandDmayhavenegativelyinfluenced theoutcomeofthisstudy. Afterthela mivudineinterruptionsatra nsientincreaseinproliferationcapacityand IFNγproductionbyThcellswaso bserved,whichindicatesupregulationofaTh1 response. Furthermore,inparallelwithi ncreasedviralrepli cationandpresu mably strongerantigenexpressionbyhepatocytes,duringthe firstlamivudineinterruption thei ntrahepatic fractionofCD8+Tcellsi ncreased.Anincremento fCD8+Tcells intheliverhas been correlatedtorespo nsetoI FNαtherapy 28.Thusalthough IVItransientlysti mulatestheH BVspecificimmunerespo nsivenesso fTcells,the magnitudeoftheresponseisapparentlyinsufficienttocauseasustainedvirological effectinthemajorityofpatients. In2patientsatthee ndofthestudytheviralloadhadsta bilizedatasig nificantly reducedlevelascomparedtobaseline.Comparedtotherestofthestudypopulation, inthesepatientsIVIdidnotinducemoreexplicitproliferationofThcellsorahigher frequencyofIFNγproducingcellsafterstimulationwithHBcAginvitro.However,in bothpatie ntsarelativelystro ngriseo fthe fractionintrahepaticCD8+Tcellswas

89 CHAPTER7

detected,aswellashepatitisactivitythatappearedtoberelatedtowithdrawing lamivudinetherapy.Inoneofthem(patient14)theflarecoincidedwithtransientHBV DNAsuppression.ThismaysuggestthatinthesepatientsIVIwasabletoinducean intrahepaticanti viralimmuneresponsethatcontributedtopartialimmunecontrol overthevirusinfection. Withrespecttosafetyandtolerability ,therewasnodif ferencewithpreviousstudies usinglamivudineincombinationwithPeg IFNα 7.Nodangerousfl ares(ALT >10x upperlimitofnormal)orfl aresleadingtoliverfailurewereencounteredafter lamivudineinterruption.Also,despitethepotentialris kthatlamivudineinterruptions mayinduceviralresistance,theemergenceof YMDDmutantsduring IVI,didnot exceedthefrequencyobservedafterlamivudinemonotherapyfor12months4,29. Inconclusion,inthisprospectivepilotstudy IVIwasabletotransientlystimulatethe HBVspecificimmuneresponsivenessof Tcells,butthemagnitudeoftheresponse wasinsufficienttocauseabene ficialvirologicalef fect,comparedtocurrentlyused treatmentstrategies.Priortoinitiatingalarger,controlledstudy,factorssuchasthe potencyofvirussuppressionbeforewithdrawal,thedurationofnucleosideanalogue withdrawalorthenumberofwithdrawals,mayhavetobereconsideredtooptimi ze thestudydesign.

90 CHAPTER7CHAPTER7IVICLINICALSTUDY

References 1. KaneM.GlobalprogrammeforcontrolofhepatitisBinfection.Vaccine1995;13Suppl1:S479. 2. WrightTL,LauJY.ClinicalaspectsofhepatitisBvirusinfection.Lancet1993;342:13404. 3. NiederauC,HeintgesT,LangeS,etal.LongtermfollowupofHBeAgpositivepatientstreatedwith interferonalfaforchronichepatitisB.NEnglJMed1996;334:14227. 4. LaiCL,ChienRN,LeungNW,etal.AoneyeartrialoflamivudineforchronichepatitisB.AsiaHepatitis LamivudineStudyGroup.NEnglJMed1998;339:618. 5. AngusP,VaughanR,XiongS,etal.Resistancetoadefovirdipivoxiltherapyassociatedwiththeselection ofanovelmutationintheHBVpolymerase.Gastroenterology2003;125:2927. 6. vanZonneveldM,HonkoopP,HansenBE,etal.Longtermfollowupofalphainterferontreatmentof patientswithchronichepatitisB.Hepatology2004;39:80410. 7. JanssenHL,vanZonneveldM,SenturkH,etal.Pegylatedinterferonalfa2baloneorincombinationwith lamivudineforHBeAgpositivechronichepatitisB:arandomisedtrial.Lancet2005;365:1239. 8. SprengersD,JanssenHL.ImmunomodulatorytherapyforchronichepatitisBvirusinfection.FundamClin Pharmacol2005;19:1726. 9. PolS,NalpasB,DrissF,etal.EfficacyandlimitationsofaspecificimmunotherapyinchronichepatitisB. JHepatol2001;34:91721. 10. ManciniBourgineM,FontaineH,BrechotC,PolS,MichelML.ImmunogenicityofahepatitisBDNA vaccineadministeredtochronicHBVcarriers.Vaccine2005. 11. MichelML,PolS,BrechotC,TiollaisP.ImmunotherapyofchronichepatitisBbyantiHBVvaccine:from presenttofuture.Vaccine2001;19:23959. 12. RehermannB,FowlerP,SidneyJ,etal.ThecytotoxicTlymphocyteresponsetomultiplehepatitisBvirus polymeraseepitopesduringandafteracuteviralhepatitis.JExpMed1995;181:104758. 13. OritaniK,KincadePW,ZhangC,TomiyamaY,MatsuzawaY.TypeIinterferonsandlimitin:acomparison ofstructures,receptors,andfunctions.CytokineGrowthFactorRev2001;12:33748. 14. SenGC.Virusesandinterferons.AnnuRevMicrobiol2001;55:25581. 15. BonvinM,AchermannF,GreeveI,etal.InterferoninducibleexpressionofAPOBEC3editingenzymesin humanhepatocytesandinhibitionofhepatitisBvirusreplication.Hepatology2006;43:13641374. 16. SprengersD,JanssenHL,KwekkeboomJ,NiestersHG,deManRA,SchalmSW.Invivoimmunization followingvirussuppression:anovelapproachforinducingimmunecontrolinchronichepatitisB.JViral Hepat2003;10:79. 17. PasSD,FriesE,DeManRA,OsterhausAD,NiestersHG.Developmentofaquantitativerealtime detectionassayforhepatitisBvirusDNAandcomparisonwithtwocommercialassays.JClinMicrobiol 2000;38:2897901. 18. StuyverL,DeGendtS,VanGeytC,etal.AnewgenotypeofhepatitisBvirus:completegenomeand phylogeneticrelatedness.JGenVirol2000;81:6774. 19. NorderH,CourouceAM,MagniusLO.Completegenomes,phylogeneticrelatedness,andstructural proteinsofsixstrainsofthehepatitisBvirus,fourofwhichrepresenttwonewgenotypes.Virology1994; 198:489503. 20. IshakK,BaptistaA,BianchiL,etal.Histologicalgradingandstagingofchronichepatitis.JHepatol1995; 22:6969. 21. LautenschlagerI,HockerstedtK,HayryP.Fineneedleaspirationbiopsyinthemonitoringofliver allografts.TransplInt1991;4:5461. 22. SprengersD,vanderMolenRG,KustersJG,etal.Flowcytometryoffineneedleaspirationbiopsies:a newmethodtomonitortheintrahepaticimmunologicalenvironmentinchronicviralhepatitis.JViralHepat 2005;12:50712. 23. LauGK,SuriD,LiangR,etal.ResolutionofchronichepatitisBandantiHBsseroconversioninhumans byadoptivetransferofimmunitytohepatitisBcoreantigen.Gastroenterology2002;122:61424. 24. WherryEJ,BlattmanJN,MuraliKrishnaK,vanderMostR,AhmedR.ViralpersistencealtersCD8Tcell immunodominanceandtissuedistributionandresultsindistinctstagesoffunctionalimpairment.JVirol 2003;77:491127. 25. LohrHF,KrugS,HerrW,etal.QuantitativeandfunctionalanalysisofcorespecificThelpercellandCTL activitiesinacuteandchronichepatitisB.Liver1998;18:40513. 26. BoniC,BertolettiA,PennaA,etal.LamivudinetreatmentcanrestoreTcellresponsivenessinchronic hepatitisB.JClinInvest1998;102:96875. 27. MarinosG,NaoumovNV,WilliamsR.Impactofcompleteinhibitionofviralreplicationonthecellular immuneresponseinchronichepatitisBvirusinfection.Hepatology1996;24:9915. 28. TangTJ,KwekkeboomJ,ManchamS,etal.IntrahepaticCD8+Tlymphocyteresponseisimportantfor therapyinducedviralclearanceinchronichepatitisBinfection.JHepatol2005;43:4552.

91 CHAPTER7

29. SantantonioT,MazzolaM,IacovazziT,MigliettaA,GuastadisegniA,PastoreG.Longtermfollowupof patientswithantiHBe/HBVDNApositivechronichepatitisBtreatedfor12monthswithlamivudine.J Hepatol2000;32:3006.

92 CHAPTER 8

Induction of CD4+ CD25+ regulatory T-cells associated with nonresponsetopegylatedInterferonαtherapyforchronic hepatitisBvirusinfection

D.Sprengers1,J.N.Stoop1,R.S.Binda1,J.G.Kusters1,B.Haagmans2,P.Carotenuto2, A.Artsen2,A.D.M.E.Osterhaus2,R.G.vanderMolen1*H.L.A.Janssen1*. *Equalcontribution

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedical Center,Rotterdam,TheNetherlands 2 Dept.ofVirology,ErasmusMCUniversityMedicalCenter,Rotterdam, TheNetherlands

Submitted CHAPTER8

Abstract

Littleisknownaboutwhytreatmentwithinterferonalpha(IFNα)leadstoaresponse inonlyaminorityofpatientswithchronicHBV . Itwasrecentlyshownthatinthese patientsCD4 +CD25+regulatoryT cells(T reg)cansuppresstheHBV specific immuneresponse. Weaimedtoin vestigatewhetherinnonresponderstoIFNα therapyT regcontributetotreatmentfailurebydownre gulatingtheHBV specific Tcellresponses.Fourteenpatientsrecei vedpe gylatedIFNαmonotherapyfor 52 weeksandwerefollowedupfor26weeks.Comparedtononrespondersresponders displayedanincreasedHBV specific Thcellproliferation. Whereastherewasno differenceinthefre quenciesofCD4 +CD25+Tregbetweenrespondersandnon respondersatthestartoftreatment ,duringandaftertherapyinrespondersthese frequenciesdecreased ,andinnonresponderstherewasasi gnificantincreasein thefrequencyofCD4+CD25+Tregduringtreatment.Incontrasttotheresponders , thenonrespondersshowedasi gnificantincreaseinfre quencyofIL 10producing cells.T regdepletionresultedinincreasedproliferationcapacityandincreased frequenciesofHBV specific INFγproducingcells,butdidnotaf fectthefre quency ofIL10producingcellsmeasuredduringthecourseofthetreatmentInconclusion , thisstudyindicatesthattheremaybeanimportantroleforregulatoryTcells,inHBV persistenceduringandafterPegIFNαtherapy.

94 CHAPTER8CHAPTER8REGULATORYTCELLSDURINGIFNTHERAPY

Introduction

ChronicinfectionwithhepatitisBvirus(HBV)ischaracterizedbyinfl ammatoryliver diseaseofvariableseverityandisamajorcauseofliverfailureandhepatocellular cancerworldwide1,2.HBVisanoncytopathicvirus,andliverinjuryismainlymediated bythehostimmuneresponseagainstvirusinfectedlivercells3.Therefore,inchronic hepatitisBinfection(CHB)effectivetherapyisneededtostopviralreplicationand progressionofliverdamage. Currently, interferonalpha(IFNα),adrugthatmodulatestheantiviralimmune response,representsthefirsttreatmentofchoiceinCHB4.However,inonly2030% ofCHBpatientsconventionalIFNαtherapyleadstosustainedvirologicalresponse 5,6andtherecentintroductionofpegylatedIFNα,adrugwithaprolongedhalflife, hasnotchangedthepicturethatthemajorityoftreatedpatientsdoesnotrespondto therapy7. Previousstudieshavedefi nedtwo Th elpercellsubsets,Th1andTh2,whichare characterizedbydistinctandmutuallyexclusivepatternsofcytokineproduction anddifferentfunctions 8.Th1cellsproduceIFNγ,IL2andpromotecellularimmune reactions,whileTh2cellsproduceIL4,5,andIL13,andenhancehumoralimmune response8.ResolutionofacuteHBVinfectionhasbeenassociatedwithavigorous polyclonalandmultispecifi c Th1response 9,whereasinthebloodofchronically infectedpatientstheHBV specificTh1response isweak,antigenicallyrestrictedor undetectable 10,11.Peripheral Tc ellscontainanimmunoregulatorysubpopulation whichexpressesCD4,CD25(theIL2receptorαchain)andCD45RO,andhavea continuousintracellularexpressionofthecytotoxicTlymphocyteassociatedantigen 4(CTLA4).Theseregulatory Tc ells(T reg)arecapableofinhibitingtheef fector functionsofCD4+,CD8+andnaturalkillerTcells1215.Inarecentstudyitwasshown thatpatientswithachronicHBVinfectionhaveahigherpercentageof Tregintheir peripheralbloodcomparedtohealthycontrolsandindividualswitharesolvedHBV infection16.TheseTregwereabletosuppresstheHBVspecificimmuneresponsein adosedependentmanner.Althoughthemechanismofthissuppressiveeffectisnot clear,studiesinotherchronicviralinfectionshaveshownthatuponstimulation Treg maycontributetopersistentviralinfectionbymodulationofthevirusspecifi c Th1 response17. LittleisknownaboutwhytreatmentwithpegylatedIFNleadstoaresponseinonly aminorityofpatients.Elucidatingthemechanismresponsiblefortreatmentfailure mayresultinimprovedcontrolofthevirusinfectionandbettertreatmentstrategies. Therefore,inthisstudy,weaimedtoanalyzelongitudinallythevirusspecifi cThcell responsesandcytokinepatternsduringandaftertreatmentofpatientswithCHB withPegIFNα2bmonotherapy.W ehypothesizedthatinnonresponderstoIFNα therapyTregcontributetotreatmentfailurebydownregulatingtheHBVspecificTh1 responses.

95 CHAPTER 8 REGULA

Patients and methods

Patients Patient material used for this study was derived from individuals that participated in a multicenter, randomized, double-blind study carried out to compare the efficacy of pegylated interferon-alpha 2b (Peg-IFN) monotherapy to a combination regimen with lamivudine for treatment of patients with HBeAg positive chronic hepatitis B 7. All patients were HBeAg positive, had alanine amino transferase (ALT) levels twice the upper limit of normal, had not received antiviral therapy within 6 months prior to enrollment and displayed no serious co morbidity. Patients were treated for 32 weeks with 100μg Peg-IFN weekly and with 50μg Peg-IFN weekly for another 20 weeks (total 52 weeks) after which they were followed-up for 26 weeks. Response to therapy was defined as HBeAg-loss at the end of follow-up. For the current study we used material from all locally enrolled patients that retrospectively had received Peg-IFN monotherapy, and of whom material was obtained after signing informed consent (n=14).

Isolation of the PBMCs and Flow Cytometric Analysis At the start of therapy (t=0), 8 weeks later (t=8), at the end of therapy (t=52) and at the end of follow-up (t=78) PBMCs from patients were obtained by ficoll separation (Ficoll-PaqueTM plus, Amersham Biosciences, Buckinghamshire, UK). The PBMCs were immediately frozen in medium containing 10% DMSO and stored at –135°C until further use. PBMC from different timepoints were tested simultaneously to avoid interassay variations. Flow cytometric analysis was performed on the stored samples using fluorochrome conjugated antibodies specific for the surface markers CD4, CD45RO and CD25 diluted in PBS/ 0.3% bovine serum albumin. The cells were fixed by incubation with intraprep reagent 1 and permeabilized by incubation with intraprep reagent 2 (Beckman-Coulter, Marseille, France). Anti-CTLA-4 antibody was added during permeabilization. The following antibodies were used: anti-CD4- PerCP-Cy5.5 (SK3) (Pharmingen, San Diego, CA), anti-CD45RO-APC (UCHL1) (Becton Dickinson, San Jose, CA), anti-CD25-FITC (2A3) (Becton Dickinson), anti- CTLA-4-PE (BNI3) (Immunotech, Marseille, France). For the CD45RO, CD25 and CTLA-4 antibodies, isotype matched control antibodies were used to determine the level of background staining. After staining the cells were analyzed using a four- color cytometer (FACScaliburTM, CELLQuest ProTM software, Beckton Dickinson). The FoxP3 antibody (clone PCH101, eBiosciences, San Diego, Ca) staining was performed according the manufacturers instructions. Briefly, cells surface markers were stained with anti CD25-PE (M-A251) (Pharmingen) anti-CD4-PerCP-Cy5.5 (SK3) (Pharmingen). The cells were fixed and permeabilized with Fix/perm buffer and permeabilization buffer (eBiosciences) anti-FoxP3-APC was added during permeabilization. To determine the level of background staining, for anti-CD25-PE and anti-FoxP3-APC isotype matched control antibodies were used.

96 CHAPTER 8 REGULATORY T-CELLS DURING IFN THERAPY

CD4+CD25+ T cell isolation Thawed PBMCs obtained at t=0, t=8, t=52 and t=78 were used for CD4+CD25+ T cell isolation. CD4+ T cells were isolated from PBMCs by negative selection using the untouched CD4+ T cell isolation kit (Miltenyi Biotec, Bergisch Gladbach, Germany). CD4+CD25+ T cells were isolated from CD4+ T cells using anti CD25-microbeads (Miltenyi biotec). The isolations were performed according to manufacturer’s instructions. The CD4- and the CD4+CD25- fraction were pooled and were used as CD25 (Treg)-depleted responder cells. Purity of the cell fractions was determined by flow cytometry analysis with antibodies against CD3, CD4 and CD25. The following antibodies were used: anti-CD3-FITC (clone UCHT1, Immunotech), anti-CD4- PerCP-Cy5.5 (clone SK3, Becton Dickinson) and anti-CD25-PE (clone M-A251, Pharmingen). An isotype matched control antibody was used to determine the level of background staining for the anti-CD25-PE antibody. The CD4+CD25+ Treg purification method resulted in a Treg fraction containing more than 90% pure CD4+CD25+ Treg and a Treg-depleted cell fraction containing all other cell types present in PBMCs.

Proliferation assay Th-cell proliferation was determined as reported previously 18. Isolated PBMCs and the pooled CD25-depleted cells of all time points investigated were cultured in triplicate in a concentration of 1X105 cells per well in 100 μl RPMI 1640 (Bio Whittaker, Verviers, Belgium) containing 5% pooled human serum (Dept. of Immunohematology and Bloodbank, Leiden University Medical Center, Leiden, The Netherlands) and penicillin/streptomycin (Gibco, Paisley, UK). The cells were stimulated with 1 μg/ml HBV core antigen (HBcAg) (a kind gift by M van Roosmalen, Biomerieux, Boxtel, The Netherlands), 5 μg/ml Phytohemagglutin (Murex, Paris, France), or not stimulated and cultured for 6 days. After 5 days of incubation the cells were pulsed with 0.25 μCi/well of [3H]thymidine (Radiochemical Centre, Amersham, Little Chalfont, UK). The cells were harvested 16 hours later. Proliferation, was determined by liquid scintillation counting of the harvested cells and expressed as stimulation index (SI). The SI was calculated from the counts per minute found with antigen divided by that found without antigen.

Elispot assay Detection of IFNγ (U-CyTech, Utrecht, The Netherlands) and IL-10 (Sanquin, Amsterdam, The Netherlands) was performed using ELISPOT kits according to the manufacturer’s instructions. Of all time points investigated, cultures of PBMCs and the pooled CD25-depleted cells of were established in triplicate on round bottom 96-wells plates (Corning Inc., New york, USA), in a concentration of 1X105 cells per well in 100 μl RPMI 1640 (Bio Whittaker) containing 5% pooled human serum (Leiden University Medical Center) and penicillin/streptomycin (Gibco). The cells were stimulated with 1 μg/ml HBV core antigen (HBcAg) (M. van Roosmalen, Biomerieux), 5 μg/ml Phytohemagglutin (Murex) (positive control) or medium only (negative control) for 24 hours at 37 degrees Celsius, after which they were transferred into flat bottom,

97 CHAPTER8

antiIFNγorantiIL10coated96wellsplates(Nunc A/S,Roskilde,Denmarkand Millipore,Molsheim,France,respectively)foranother24hoursincubationperiod. Cytokinespotswerevisualisedbybiotinlabelledantibodiesandtheirnumberswere establishedbyusingaBioreader3000fromBioSys(Karben,Germany). Ar esponse wasconsideredpositiveifthenumberofspotswasequaltoorgreaterthan2.5 timesthebackground,andthereportedHBcAgspecificdataisthenumberofspots countedminusthebackgroundlevel.

Statistics Treg flowcytometrydata,cytokineprofi lesandproliferationcapacityofPBMC obtainedfromnonrespondersandresponderstoantiviraltherapywerecompared usingtheMannWhitneyUtest.LongitudinalanalysisoffrequenciesofTreg,cytokine profilesandproliferationcapacityduringthestudyperiod,wasperformedwiththe Wilcoxonmatchedpairssignedranksumtest.FortheseanalysesSPSS1 1.5for Windows(SPSS,Chicago,IL)wasused. Whereapplicabledataarereportedas mean±standarderrorofthemean(SEM).

Results

Patients TableIshowsthepatientcharacteristics.Attheendoffollowup(t=78)8patientshad remainedHBeAgpositive(nonresponders),and6patientshadsustainedlossof HBeAg(responders;43%).Atthestartoftreatment(t=0),the8nonrespondershada medianALTof102IU/l(range47394)versus168IU/l(range72236)inresponders, andthemedianviralloadmeasured2.3 x109cp/ml(range1.6 x1087.1x109)versus 1.1x109cp/ml(range1. 7x1085.7x109),respectively .These differenceswerenot significant.At theendoftherapyandattheendoffollowupHB VDNAl evels were1.8 x108cp/ml(1.4 x1034.7x108)and5.3 x108cp/ml(3.4 x1054.9x109)innon responders,vs.1.0x103cp/ml(4802.9x104)and473cp/ml(<373970)inresponders, respectively.

HBVspecificThelpercellproliferation Ap roliferationassaywasperformedtomeasuretheimmuneresponsivenessof Th cellstoHBcAglongitudinallyduringthecourseofthetreatment. Whereastherewas considerablevariationbetweentheresponders,thenonrespondersshowedasimilar patternofresponsecharacteri zedbyadecreasingproliferationcapacityof Thcells duringPegIFNαtherapy,whichappearedtorestoreinthefollowupperiod(fi g1). Atthestartoftherapytherewasnosignifi cantdifferencebetweenrespondersand nonresponders(meanSI =4.83±0.63and4.43 ±0.38,respectively).However,in nonrespondersat8and52weeksthemeanproliferationcapacityofThcellshad decreasedby35 %and76%,respectivelycomparedtot =0(p <0.05),whereas5 of6respondersshowedaprofoundincreaseinthe Thcellproliferativeresponseto HBcAg(SI>2xbaselinelevel)atoneormoretimepointsanalyzed.

98 CHAPTER8CHAPTER8REGULATORYTCELLSDURINGIFNTHERAPY

Table I. Patient characteristics

Patients Sex Age Race ALT HBV-DNA Response to (case) (U/l) (geq/ml) therapy t=0 t=0 1 Male 58 Cauc. 78 1.6x109 - 2 Male 47 Asian 125 1.9x108 - 3 Male 32 Asian 47 1.6x108 - 4 Male 27 Cauc. 70 3.0x109 - 5 Male 28 Cauc. 325 7.1x109 - 6 Male 49 Cauc. 394 5.7x109 - 7 Male 38 Cauc. 79 7.9x109 - 8 Female 21 Cauc. 160 2.2x108 - 9 Male 35 Cauc. 151 5.2x109 + 10 Male 41 Asian 131 1.2x109 + 11 Male 43 Cauc. 185 5.7x109 + 12 Male 41 Asian 236 4.4x108 + 13 Male 29 Cauc. 203 9.9x108 + 14 Female 29 Asian 72 1.7x108 +

All 14 patients were HBeAg positive at inclusion and were treated with PEG-IFNα for 52 weeks. At the end of therapy 6 patients (case 9 – 14) had serocoverted to HBeAg-negative and this seroconversion was sustained at the end of a follow-up period of 26 weeks. There were 8 non-responders (HBeAg positive, case 1-8) at end of therapy. t = 0 is start of therapy

20 x

e 16 d n I

n o

i 12 t Responders a l u Non-responders m

i 8 t S

4

0 t=0 t=8 t=52 t=78

Time point

Fig1.Meanproliferationcapacity(±sem)of ThcellsofpatientswithchronicHBV , obtainedatdif ferenttimepointsduringPEGIFNαtherapy ,afterstimulationwith HBcAg.The timepointsanalyzedwere:t=0,t=8,t=52andt=78weeks.Innon responders,comparedtot=0att=8andt=52thereductioninstimulationindex wassignificant(p=0.016atbothtimepoints). At8and52weeksoftreatmentthe differencesbetweenrespondersandnonrespondersshowedatrendtowards significance(p=0.07andp=0.10,respectively).

99 CHAPTER8

LongitudinalanalysisofIFNγproducingcellsinresponsetoHBcAg ToanalyzethekineticsofantiviralTh1activity,thefrequencyofIFNγproducingcells afterinvitrostimulationwithHBcAgwasdeterminedlongitudinallyduringtherapy. Therewasawidevariationbetweenindividualpatients,andnotatanytimepoint analyzedtherewasasignificantdifferenceinfrequenciesofIFNγproducingThcells betweenrespondersandnonresponders(fig2).AttheendofPEGIFNαtherapyin 6of8nonrespondersandin4of6respondersthemeannumberofIFNγproducing cellsafterstimulationwithHBcAg,haddecreased.Comparedtot=0,atthistimepoint inthenonrespondersandresponderstherewasameanreductionof39%and72% (p=0.18andp=0.43),respectively. C

M 16 B P

5 14 0 1

x 12 1

/

s 10 l l

e Responders c 8 g Non-responders n i

c 6 u d

o 4 r p

γ 2 N F I 0 t=0 t=8 t=52 t=78

Time point Fig2.Meanfrequencies(±sem)ofIFNγproducingThcellsattreatmentweek0,8,52and78, accordingtotreatmentresponse.

FrequenciesofCD4+CD25+regulatoryTcellsduringPEGIFNαtherapy AtalltimepointsinvestigatedwedeterminedthefrequenciesofCD4+CD25+Treg. BaseduponseveralrecentstudieswhichshowedthattheCD4+CD25+cellswithhigh expressionofCD25areTreg12,16,19,CD45ROandCTLA4wereusedasadditional markers in our flow cytometry experiments to identify this cell population.Typical dataasobtainedbyflowcytometryofPBMCsfromarepresentativeresponderand nonresponder,areshowninfigure3a.Figure3bshowsthemeanfrequenciesof CD4+CD25+Treginrespondersandnonrespondersduringtreatment.Atbaseline respondersandnonrespondershadacomparablefrequencyofCD4+CD25+Treg (2.21±0.10vs2.23±0.19respectively).Attheendoftherapyinnonrespondersthe frequencyhadincreasedby74%toameanof3.6%±0.18(p=0.008),whereasin respondersithaddecreasedby20%toameanof1.75%±0.09(p=0.031;responders vs. nonresponders p =0.008).At the end of followup in nonresponders the frequencyCD4+CD25+Tregreturnedtothebaselinelevel,whereasinrespondersit furtherdecreasedtobelowbaseline(respondersvs.nonrespondersp=0.074). CD4+CD25+Tregexpresstheforkhead/wingedhelixfamilyproteinFoxp3,whichisa keyregulatorforthedevelopmentandfunctionofTreg,andcurrentlythemostspecific marker for this cell population 2022.The recent availability of an adequateFoxp3

100 CHAPTER8CHAPTER8REGULATORYTCELLSDURINGIFNTHERAPY

A Non Responder

1.4% 2.7% 3.2% 2.5% e P P C CD25 Fitc IgG Fitc 4 r - e A CD45RO APC P L

Responder T 4 C D 3.2% 1.7%3.0%

C 1.6%

CD25 Fitc IgG Fitc CD45RO APC t = 0 t = 8 t = 52 t =78

Timepoint B 4 * s l l e

c 3

+ 4

D Responders C

+

f 2

o x Non-responders g e r T 1 %

0 t=0 t=8 t=52 t=78

Time point

* p = 0.008 compared to t=0 + p = 0.031 compared to t=0 x p = 0.028 compared to t=0 Fig3.PercentageofTreg(CD4,CD25,CD45ROandCTLA4positivecells)withintheCD4 + cellfraction,determinedbyFACSstainingonPBMCof8nonrespondersand6respondersto PEGIFNαtherapy.Thetimepointsanalyzedweretreatmentweek0,8,52and78. 3a)Flowcytometrystainingexperimentfromarepresentativeresponder(case10)andnon responder(case4)toPegIFNαtherapy,duringthecourseofthestudy.Antibodieswereused againstCD4,CD25,CD45ROandCTLA4.Thegateswerepositionedusingisotypematched controlantibodiesasshowninthetwodotblotsontheleft.CD4 +CD25+cellswereanalyzed forCTLA4andCD45ROexpression,andthenumberinthetheupperrightquadrantofde fourdotblotsontherightindicatesthepercentageCTLA4+CD45RO+CD4+CD25+cellsof totalCD4+cells(Treg). 3b)Meanfrequenciesof Treg(±sem)duringthestudy .Comparedtothebaselinelevel,in nonresponderstherewasasignificantincreaseinthefrequencyofTregattheendoftherapy, whereasinresponderstherewasasignificantdecrease.Attheendoftherapythefrequency ofTregwashigherinnonrespondersthanresponders(p=0.008).Att=78innonresponders thefrequencyhadreturnedtobaselinelevel,whereasinrespondersitcontinuedtodecrease (respondersvs.nonrespondersp=0.074)

101 CHAPTER8

Fig3c)Foxp3expressionbyCD4+CD25+CTLA4+cellsinarepresentative untreatedchronicHBVpatient. Thenumberintherightupperquadrantoftheright dotblotindicatesthepercentageFoxp3+cellsoftotalCD4+CD25+CTLA4+cells.

antibodyenabledustovalidateourTregstaining,bydetermininginaseparategroup of5untreatedchronicHBVpatientstheFoxp3expressionbyCD4+CD25+CTLA4+ cells.Almost allCD4+CD25+CTLA4+cellsexpressedtheFoxp3protein(mean percentageFoxp3+/CD4+CD25+CTLA4+cells:98%±0.46),whichindicatesthat thecombinationofcellsurfacemarkersusedtodeterminethe Tregfrequenciesin thestudypopulation,indeedmostlikelyidentifiesasuppressiveCD4+CD25+Tcell population.Figure3cshowstheflowcytometrydataofarepresentativepatient.

LongitudinalanalysisofIL10producingcellsinresponsetoHBcAg Previously, ithasbeensuggestedthatinchronicHBVpatientstreatedwiththe combinationofribavirinandIFN α,IL10cytokineproductionisassociatedwith responsetotherapy 23.Therefore,inourpatientpopulationwelongitudinallystudied thefrequencyofIL10producingThcellsafterstimulationwithHBcAg in vitro. Therewasawidevariationbetweenindividualpatientsofbothpatientgroups. At notimepointanaly zedtherewasasignifi cantdifferencebetweenrespondersand nonresponders(fig4).However,incontrasttotheresponders,duringtreatmentthe nonrespondersshowedagradualincreaseinfrequencyofIL10producingcellsfrom ameanof22. 7±4.0/1x10 5PBMCatt =0toameanof48±6. 7/1x105PBMCat t=52weeks(p =0.016).This numberreturnedtothebaselinelevelaftercessation ofthetherapy.InseveralstudiesIL10hasbeenimplicatedasapotentialmediator ofsuppressionoftheimmuneresponsebyCD4+CD25+ Treg 17,24 .Although, in respondersandnonrespondersthetrendsofIL10producingcellsandofCD4+ CD25+Tregweresimilar ,therewasnosignifi cantcorrelationbetweenthesetwo parameters(datanotshown).

HBVspecificimmuneresponseafterCD4+CD25+regulatoryTcelldepletion Infourpatients(2responders,2nonresponders)suffi cientcellswereobtainedto

102 

120 PBMC 5 100

80 Responders 60 *** Non-responders 40

producing cells 1x10 / 20 10 10 IL- IL- 0 t=0 t=8t=8 t=52t=52 t=78 t=78

Time point

Non-responders: * p = 0.016 compared to t=0  

                            

14 * 12 10 * 8 6 4 2 0 Stimulation Index Stimulation t=0 t=8 t=52 t=78   60 * 50        40 *

PBMC 30 *       5 5 20        producing cells / producing

1x10 10 γ        0 IFN t=0 t=8 t=52 t=78     

120        100  80

PBMC  5 5 60          40 1x10 20        

IL-10 cells / producing 0       t=0 t=8 t=52 t=78 Time point   PBMC PBMC after CD4+ CD25+ cell depletion 

 CHAPTER8

Discussion

PegIFNαtherapyleadstosustainedvirologicalresponseinaminorityofchronicHBV infectedpatients7.Elucidatingthemechanismresponsiblefortreatmentfailuremay resultinimprovedcontrolofthevirusinfectionandbettertreatmentstrategies.Inthis studywehaveinvestigatedtheHBVspecificThcellresponsesinperipheralbloodof HBeAgpositiveCHBpatientstreatedwithPEGIFNα.Sinceitwasrecentlyshown thatpatientswithchronicHBVinfectionhaveahigherpercentageofCD4+CD25+ Tregintheirperipheralbloodthanindividualswitharesolvedinfection,andthat theseTregwerecapableofinhibitingtheHBVspecificimmuneresponse16,wehave investigatedtheroleofthis Tcellpopulationintheoutcomeoftherapy .Theresults showthatduringtherapy,inthemajorityofrespondersthereisincreasedproliferation ofThcellsafterstimulationwithHBcAg,whereasinnonrespondersthereisaclear decrease.Previously,ithasbeendescribedthatresponderstoconventionalIFNα displayasimilarincreasedThcellproliferationcapacity25whichmayreflectincreased Thcellreactivitytothevirus.Incontrasttoseveralreportsdescribingincreased IFNγproductioninresponderstoIFNαtherapyonly 25,26 ,wefoundadecreasein thefrequencyofIFNγproducingcellsduringtherapyinbothrespondersandnon responders,toasimilarextend.Thisdiscrepancymaybeexplainedbydifferencesin timepointsinvestigatedorbymethodologicaldifferences,sincepreviouslyIFNγwas measuredinsupernatants25,26whereaswequantifiedthefrequencyIFNγproducing cells.OnecouldhypothesizethatafterinvitrostimulationwithHBcAg,theThcellsof respondersaremorepotentIFNγproducersthanthoseofnonresponders. Atthestartoftreatmenttherewasnodif ferenceinthefrequenciesofCD4+CD25+ Tregbetweenrespondersandnonresponders.However ,duringPEGIFNαtherapy innonresponderstherewasasignificantincreaseinthefrequencyofCD4+CD25+ Treg,whereasinrespondersthisfrequencyhadsignifi cantlydecreasedbothatthe endoftherapyandfollowup.NocorrelationwasfoundbetweenHBVDNA levels orALTandthepercentageof Treg(datanotshown).TregexpresstheFoxp3gene, whichisimportantinthedevelopmentandsuppressivefunctionofCD4+CD25+ Treg21,22,andweandothers 16showedthattheyarecapableofinhibitingtheHBV specificimmuneresponse.Therefore,onecouldhypothezise thatanincrementin TregfrequencyduringPEGIFNαtherapymaynegativelyinfluenceThcellreactivity tothevirus,andcontributetononresponsetotreatment.CytotoxicTcellresponses aresubjectedtonegativecontrolbyCD4+CD25+ Treg 27.Also, inmicevaccine inducedvirusspecific Tcellresponseswereshowntobesuppressedbyanother majorregulatoryTcellpopulationthathasbeenimplicatedinthecontrolofspecifi c Tcellimmunity,namelyCD4+IL10+ Tregulatorycells(Tr1cells) 28,29.IL10could beinstrumentalin Tr1mediatedsuppressionof Tcellproliferationandcyto kine productioninacellcellcontactindependentmanner 30.ThesemouseTr1cells,but notCD4+CD25+ Treg,wereupregulatedundertheinfl uenceofIFNα β 28.Also, humanTr1cellscanbegeneratedinvitroundertheinfluenceofexogenousIFNα31. Inourstudy,duringthecourseofPEGIFNαtherapywefoundthatincontrastto

104 CHAPTER8CHAPTER8REGULATORYTCELLSDURINGIFNTHERAPY

theresponders,thenonrespondersshowedasignifi cantincreaseinfrequency ofIL10producingcells.IL10isimportantinblockingproinfl ammatorycytokine production,costimulation,MHCclassIIexpression,andchemokinesecretion32,and increasedIL10productionhaspreviouslybeendescribedinchronicHBV patients notrespondingtoribavirinIFNαcombinationtherapy 23.Thisobservationsuggests adissociationinIL10productioninrelationtoresponsetotherapy.Wewereunable toshowacorrelationbetweenthefrequenciesofCD4+CD25+ TregandIL10 producingcellsintheindividualpatients. Also,despiteourfindingthatdepletionof theCD4+CD25+ Tc ellpopulationresultedinincreasedproliferationcapacityand increasedfrequenciesofINFγproducingcellsafterstimulationwithHBcAg,itdidnot significantlyaffectthefrequencyofIL10producingcellsmeasuredduringthecourse ofthetreatment. Therefore,innonresponderstheimmunomodulatoryef fectsof PEGIFNαtherapymayresultinanincrementinTregfrequencyandanupregulation ofIL10producingcells,(potentiallyIFNαinduced Tr1cells),whichtogethermay negativelyinfluenceTcellreactivitytothevirus. PreviousstudiesconductedinpatientswithchronicHBVinfectiontreatedwithdifferent treatmentregimes 25 3335,suggestthatthereisanassociationbetweenimproved HBVspecificTcellresponsivenessandsignificantreductionsinviremialevels.Inour study,innonrespondersanincrementinthefrequenciesofTregandIL10producing cellscoincidedwithareductioninviralload,whereasinrespondersbothparameters decreasedinparallelwithdecreasingviremia.Therefore,theupregulationofthese immunoregulatoryfactorsobservedinnonrespondersismostlikelynotduetoa reductioninthelevelsofHBVDNA. Inconclusion,thisstudyindicatesthattheremaybeanimportantroleforregulatory Tcells,inHBV persistenceduringandafterPegIFNαtherapy .Wefoundthatan increasingfrequencyofIL10producingcellsandCD4+CD25+Tregduringtreatment wasinverselycorrelatedtotreatmentresponse. To improveimmunemodulatory treatmentinchronicHBVinfection,futureinvestigationsshouldfocusonstrategies tomanipulatethefunctionoftheseregulatory Tcells,bymeansoftheirdepletionor blockadeofeffectorcytokinefunction.

105 CHAPTER8

References 1. KaneM.GlobalprogrammeforcontrolofhepatitisBinfection.Vaccine1995;13Suppl1:S479. 2. WrightTL,LauJY.ClinicalaspectsofhepatitisBvirusinfection.Lancet1993;342:13404. 3. RehermannB,FowlerP,SidneyJ,etal.ThecytotoxicTlymphocyteresponsetomultiplehepatitisBvirus polymeraseepitopesduringandafteracuteviralhepatitis.JExpMed1995;181:104758. 4. SprengersD,JanssenHL.ImmunomodulatorytherapyforchronichepatitisBvirusinfection.FundamClin Pharmacol2005;19:1726. 5. vanZonneveldM,HonkoopP,HansenBE,etal.Longtermfollowupofalphainterferontreatmentof patientswithchronichepatitisB.Hepatology2004;39:80410. 6. ManesisEK,HadziyannisSJ.InterferonalphatreatmentandretreatmentofhepatitisBeantigennegative chronichepatitisB.Gastroenterology2001;121:1019. 7. JanssenHL,vanZonneveldM,SenturkH,etal.Pegylatedinterferonalfa2baloneorincombinationwith lamivudineforHBeAgpositivechronichepatitisB:arandomisedtrial.Lancet2005;365:1239. 8. PaulWE,SederRA.Lymphocyteresponsesandcytokines.Cell1994;76:24151. 9. GuidottiLG,AndoK,HobbsMV,etal.CytotoxicTlymphocytesinhibithepatitisBvirusgeneexpressionby anoncytolyticmechanismintransgenicmice.ProcNatlAcadSciUSA1994;91:37648. 10. RehermannB,LauD,HoofnagleJH,ChisariFV.CytotoxicTlymphocyteresponsivenessafterresolutionof chronichepatitisBvirusinfection.JClinInvest1996;97:165565. 11. MainiMK,BoniC,LeeCK,etal. TheroleofvirusspecificCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 12. BaecherAllanC,BrownJA,FreemanGJ,HaflerDA.CD4+CD25highregulatorycellsinhumanperipheral blood.JImmunol2001;167:124553. 13. DieckmannD,PlottnerH,BerchtoldS,Berger T, SchulerG.Exvivoisolationandcharacterizationof CD4(+)CD25(+)Tcellswithregulatorypropertiesfromhumanblood.JExpMed2001;193:130310. 14. CamaraNO,SebilleF,LechlerRI.HumanCD4+CD25+regulatorycellshavemarkedandsustainedeffects onCD8+Tcellactivation.EurJImmunol2003;33:347383. 15. AzumaT,TakahashiT,KunisatoA,KitamuraT, HiraiH.HumanCD4+CD25+regulatory Tcellssuppress NKTcellfunctions.CancerRes2003;63:451620. 16. StoopJN,vanderMolenRG,BaanCC,etal.Regulatory Tc ellscontributetotheimpairedimmune responseinpatientswithchronichepatitisBvirusinfection.Hepatology2005;41:7718. 17. CabreraR,TuZ,XuY, etal.AnimmunomodulatoryroleforCD4(+)CD25(+)regulatory Tlymphocytesin hepatitisCvirusinfection.Hepatology2004;40:106271. 18. LauGK,SuriD,LiangR,etal.ResolutionofchronichepatitisBandantiHBsseroconversioninhumansby adoptivetransferofimmunitytohepatitisBcoreantigen.Gastroenterology2002;122:61424. 19. GrossmanWJ,VerbskyJW,BarchetW,ColonnaM,AtkinsonJP,LeyTJ.HumanTregulatorycellscanuse theperforinpathwaytocauseautologoustargetcelldeath.Immunity2004;21:589601. 20. HoriS,NomuraT,SakaguchiS.ControlofregulatoryTcelldevelopmentbythetranscriptionfactorFoxp3. Science2003;299:105761. 21. WalkerMR,KasprowiczDJ,GersukVH,etal.InductionofFoxP3andacquisitionofTregulatoryactivityby stimulatedhumanCD4+CD25Tcells.JClinInvest2003;112:143743. 22. WingK,SuriPayerE,RudinA.CD4+CD25+regulatory Tc ellsfrommousetoman.ScandJImmunol 2005;62:115. 23. RicoMA,QuirogaJA,SubiraD,etal.HepatitisBvirusspecificTcellproliferationandcytokinesecretionin chronichepatitisBeantibodypositivepatientstreatedwithribavirinandinterferonalpha.Hepatology2001; 33:295300. 24. MaloyKJ,SalaunL,CahillR,DouganG,SaundersNJ,PowrieF.CD4+CD25+T(R)cellssuppressinnate immunepathologythroughcytokinedependentmechanisms.JExpMed2003;197:1119. 25. LohrHF,KrugS,HerrW,etal.QuantitativeandfunctionalanalysisofcorespecificThelpercellandCTL activitiesinacuteandchronichepatitisB.Liver1998;18:40513. 26. CrampME,RossolS,ChokshiS,CarucciP ,WilliamsR,NaoumovNV.HepatitisCvirusspecifi cTcell reactivityduringinterferonandribavirintreatmentinchronichepatitisC.Gastroenterology2000;1 18:346 55. 27. FranzeseO,KennedyPT ,GehringAJ,etal.ModulationoftheCD8+T cellresponsebyCD4+CD25+ regulatoryTcellsinpatientswithhepatitisBvirusinfection.JVirol2005;79:33228. 28. DikopoulosN,BertolettiA,KrogerA,HauserH,SchirmbeckR,ReimannJ.TypeIIFNnegativelyregulates CD8+TcellresponsesthroughIL10producingCD4+Tregulatory1cells.JImmunol2005;174:99109. 29. BluestoneJA,AbbasAK.NaturalversusadaptiveregulatoryTcells.NatRevImmunol2003;3:2537. 30. LanR Y, AnsariAA, LianZX,GershwinME.Regulatory Tc ells:development,functionandrolein autoimmunity.AutoimmunRev2005;4:35163.

106 CHAPTER8CHAPTER8REGULATORYTCELLSDURINGIFNTHERAPY

31. LevingsMK,SangregorioR,GalbiatiF ,SquadroneS,deW aalMalefytR,RoncaroloMG.IFNalphaand IL10inducethedifferentiationofhumantype1Tregulatorycells.JImmunol2001;166:55309. 32. O’GarraA,VieiraPL,VieiraP,GoldfeldAE.IL10producingandnaturallyoccurringCD4+ Tregs:limiting collateraldamage.JClinInvest2004;114:13728. 33. BoniC,BertolettiA,Penna A,etal.Lamivudinetreatmentcanrestore Tc ellresponsivenessinchronic hepatitisB.JClinInvest1998;102:96875. 34. JungMC,HartmannB,GerlachJT,etal.Virusspecificlymphokineproductiondiffersquantitativelybutnot qualitativelyinacuteandchronichepatitisBinfection.Virology1999;261:16572. 35. MarinosG,NaoumovNV,WilliamsR.Impactofcompleteinhibitionofviralreplicationonthecellular immuneresponseinchronichepatitisBvirusinfection.Hepatology1996;24:9915.

107

CHAPTER 9

Discussion: the future of immunotherapy in chronic Hepatitis B virus infection

D.Sprengers1,H.L.A.Janssen1

1Dept.ofGastroenterologyandHepatology,ErasmusMCUniversityMedicalCenter, Rotterdam,TheNetherlands

DISCUSSION

Introduction

AccordingtotheW orldHealthOrganization,anestimated350millionpeople worldwidearechronicallyinfectedwithhepatitisBvirus(HBV).Followingacute infection,approximately10%ofadultsandasmanyas90%ofinfantsinfected perinatally, themostcommonrouteoftransmission,developchronicinfection. Althoughmanyoftheseindividualseventuallyclearthevirusorachieveastateof nonreplicativeinfection,prolongedchronichepatitisleadstothedevelopmentof cirrhosis,liverfailure,orhepatocellularcarcinomainupto40%ofpatientscausing over1milliondeathsannually 1.The goalsoftreatmentinchronichepatitisBvirus (CHB)infectionaresustainedviralsuppression,normalizationofserumalanine aminotransferase(AL T),andimprovementinliverhistology ,leadingtolongterm reductionoftheriskofcomplications. Twomajorclassesofantiviraltherapeuticshavebeenadoptedtotreattheinfection: drugsthatdirectlyinterferewithvirusreplicationanddrugsthatmodulateantiviral immuneresponse.Inmostcountrieslamivudine,adefovirdipivoxilandrecentlyalso entecaviraretheonlyapprovedinhibitorsofviralreplication(HBV DNApolymerase inhibitors).AlthoughtheyleadtorapidandalmostabsolutediscontinuationofHBV replication2,breakthroughofresistantHBVvariantslimitslongtermuse 3,4.Another strategyaimsattheinductionofimmunecontrolbyatreatmentcourseoflimited duration.This approach,ofwhichinterferonalpha(IFNα)therapyisthegenerally usedexample,istobepreferred.However,inonlyaminorityofpatientsconventional orpegylatedIFNα(PegIFNα)therapyleadstosustainedvirologicalresponse 5,6 . Therefore,alternativeagentswithenhancedpotencytostimulateimmunecontrol overthevirusareneeded. Thestudyofanimalmodelsandtransgenicmousemodelsabletoexpressindividual HBVgenesorreplicatetheentireviralgenomehaveclarifi edseveralaspects connectedtoHBVinfection.Furthermore,theabilitytoanalysemanyimmunological phenomenaexvivothroughdirectquantificationofdendriticcells,antigenspecificT cells,andregulatoryTcellsinhumansandchimpanzeeshasconsiderablyincreased ourknowledgeofHBVpathogenesis.Here,wewilldiscusstheserecentfi ndings, reportedbyusandothers,andtheirpotentialasatargetforfutureimmunotherapeutic strategiesinCHBinfection.

Targeting the immune response to Hepatitis B virus infection

Innate immunity Innateimmunitygenerallyplaysaroleimmediatelyafterinfectiontolimitthe spreadofthepathogenandinitiateeffi cientdevelopmentofanadaptiveimmune response.Innatehostresponsesduringtheearlyphasesofviralinfectionsare mainlycharacterizedbytheproductionoftype1interferon(IFN)α /βcytokinesand theactivationofnaturalkiller(N K)cells.Productionoftype1IFNscanbetriggered directlybyvirusreplicationthroughcellularmechanismsthatdetectthepresenceof

111 CHAPTER9

viralRNAorDNA79,whileNKcellsareactivatedbythemodulationofthequantityof majorhistocompatibilitycomplex(MHC)classImoleculesonthesurfaceofinfected cells10.Experimentaldatacollected,mainlyinanimalmodelsbutalsoinhumans11, showthatafterinoculation,HBVdoesnotimmediatelystarttoreplicateefficiently. HBVDNAandHBVantigensarenotdetectableinserumortheliveruntil4–7weeks postinfection1214.Followingthisperiod,HBVbeginsalogarithmicexpansionphase andinfectsmosthepatocytes12,1315,16.Inthreeexperimentallyinfectedchimpanzees, nocellulargeneswereactivatedwithintheliverduringthelagphaseofinfection,and theantiviralcytokinesIFNαand–βwerenottriggered17.Ithasbeenproposedthat becauseHBVreplicateswithinnucleocapsidparticles,viralreplicativeintermediates ofsinglestrandedRNAorviralDNA,generallystrongactivatorsoftypeIIFNgenes8, 9,areprotectedfromcellularrecognitionthroughtheTolllikereceptorsystem17. Suchearlyeventsaredifficulttoanalyseduringnaturalinfectioninhumans.HBV infectedpatientsaremainlydetectedafteronsetofclinicalsymptoms(nauseaand icterus),whichoccurwellafterinfection(10–12weeks)18.Therefore,thisearlyphase ofdiseaseislesssuitableforimmunotherapeuticinterventionsaimedatpreventing persistentinfection.

Initiation of the adaptive immune response Immediately after the exponential phase ofHBV expansion, chimpanzees able to controlthevirusshowatypicalacutephaseofdiseasewitharobustactivationof IFNγ,TNFα 13andmanycellulargeneslinkedtoaThelpertype1(Th1)typeof cellularresponse17.Theexperimentaldatainanimalmodelsareconsistentwiththe possibilitythattheinitialburstofIFNγandthesubsequentrapidinhibitionofHBV couldbemediatedbyNKandNKTcells13.Despitethesedata,theonlyexperimental evidence of NKcell involvement in humanHBV infection are represented by an analysis of NKcell frequencies in patients studied during the incubation phase of acutehepatitisB.Here,increasednumbersofcirculatingNKcellswereconcomitant withthepeakofHBVreplication,2–4weekspriortotheoccurrenceofHBVspecific CD8Tcells18.Adifferentpatternisobservedwhenpatientsoranimalmodelsdevelop chronicity.Inhumans,developmentofchronicityisoftenassociatedwithabsentor mildsymptomsofacutehepatitis.Inlinewiththeseclinicalobservations,neonatally infectedwoodchucksthatdevelopchronicitylackthelargecytokineproductionby NKTcellsobservedinresolvedanimals1921andfailtodevelopanefficientantiviral specificimmuneresponse. DrugsthatareabletoelicitstrongcytokineresponsesbyNKTcellsmaythereforebe candidatesforimmunomodulatorytherapyinCHBinfection.αGalactosylceramide (αGalCer),aglycolipidoriginallyextractedfrommarinesponges,isaligandofthe invariantVα14NKTcellandispresentedbyCD1dmoleculeonantigenpresenting cells(APC)22.AnimalstudieshaveshownthatuponactivationwithαGalCer,NKT cellsproducealargeamountofIFNγ,whichcanmodulatetheimmuneresponsesto autoimmunediseasesandinfections23,24.Ongoingclinicalstudiesshoulddetermine whetherαGalCerisaneffectiveimmunotherapeuticapproach,eitherasmonotherapy

112 DISCUSSION

orasvaccineadjuvant,inthetreatmentofCHB.

The role of CD4 T cells Theadaptiveimmuneresponseiscomprisedofacomplexwebofeffectorcelltypes, allofwhichplaykeyrolesindevelopmentofimmunitytoHBV.CD4Tcells,classically referred to as helperTcells, are robust producers of cytokines and are required for the efficient development of effector cytotoxicCD8Tcells and Bcell antibody production.CD8TcellsgoontoclearHBVinfectedhepatocytesthroughcytolytic and noncytolytic mechanisms 25, reducing the levels of circulating virus, while B cellantibodyproductionneutralizesfreeviralparticlesandcanprevent(re)infection 26.Therearecleardifferencesintheadaptiveimmunityofpatientswithestablished chronicorresolvedHBVinfection.Inchapter5weshowedthatsignificantCD8T cellresponsesaredetectableinthebloodandliverofsubjectswhoclearacuteHBV infection,evenafterresolutionofthedisease.HelperTcellandcytotoxicresponsesin acutelyinfectedpatientsarequantitativelystrongerthanthosefoundinpatientswith chronicinfections,whoareinsteadcharacterizedbyweakerorundetectablevirus specificTcellresponses27,28.ItislikelythatCD4Tcellhelpinducesthematuration ofafunctionallyefficientCD8Tcellresponse.Interestingly,weshowedinchapter4 thattheproportionofintrahepaticCD4TcellsincreasesinchronicHBVpatientswith improvedimmunecontroloverthevirus,emphasizingtheimportantroleforthiscell typeintheantiviralimmuneresponse. ItisthereforeareasonableassumptionthatimmunotherapyabletoelicitpotentCD4 Tcellresponses,maybeeffectiveinchronicHBVpatients.Indeed,inchronicHBV andchronichepatitisCinfectiontheefficacyofinterferonalphatherapyininducing sustained virologic response has been attributed to the efficacy in induction and maintenanceofsignificantThelpercellresponses29,30.However,recentattemptsto improveresponseratesusingvaccinescontainingHBVsurfaceantigeniccomponents tostimulateCD4Tcellreactivityinpatients,didnotinducecytokineproductionby thesecellsorHBVspecificCD8+Tcells31,32.Potentially,newvaccinationstrategies usingtheimmunodominantHBVcoreproteinasasubstratewillprovemoreeffective. Wewilldiscussthisfurtherbelow.

Cytotoxic T-cell response. AnalysisoftheHLAclassIrestrictedCD8TcellresponsetoHBVhasbeenseverely hamperedbytheinabilityofHBVtobepropagatedincellculture33.Thefirstdefinitive characterizationofCD8TcellsspecificforHBVderivedfromtheunderstandingthat thesequenceoftheprocessedviralantigenspresentedbyHLAclassImolecules couldbemimickedbysyntheticpeptides34.Thus,cytotoxicTcellsspecificforseveral viralepitopeswithincore34,envelope35,polymerase36andX37proteinsofHBVwere achievedusingsyntheticpeptides,andnotnaturallyprocessedepitopes,toexpand memorycytotoxicTlymphocytes(CTL)invitro.Thesestudiesdemonstratedthatthe magnitudeoftheHBVspecificCD8responseisstrongerinselflimitedthanchronic

113 CHAPTER9

infection34.Inchapter3weshowedthatflowcytometryofafineneedleaspiration biopsy of the liver allows easy, atraumatic and reliable analysis of lymphocytes obtainedfromtheintrahepaticcompartment38,andusingthistechniqueweshowed inchapter5thatinresolutionofacuteHBVinfectionthequantityofexvivoderived intrahepaticHBVspecificCD8Tcells correlated withHBV control and not with liver damage 39. Others have revealed that an epitope hierarchy exists within the HBVspecificCD8Tcellresponsesthatcanbealteredbyviralpersistence 40.The greatmajorityofA2+patientswithselflimitedhepatitisBrecognizetheHBc18–27, HBe183–91,HBe335–43andHBp455–63epitopes.Thecauseofimmunodominance of these sequences is likely linked to their good binding affinity to theHLAA2 molecule.AfurtherpossibleexplanationofthedominanceoftheseHLAA2restricted CD8responsesisthefindingthatsomeHLAclassIepitopesarenestedwithinCD4 Tcellepitopes.CD4helperTcellsarenecessaryforthemaintenanceoffunctional CD8Tcellsandthecovalentlinkagebetweenhelperandcytotoxicepitopeshasbeen showntobeimportantfortheinductionofCTLresponses41.Aminoacidmutations withintheoftenimmunodominant,HBc18–27epitopeabletoinhibitactivationofthe core18–27specificCD8Tcellshavebeenshowntooccurinpatientswithchronic hepatitisB42.LongitudinalanalysisofHLAA2restrictedHBVspecificCD8Tcellsin resolvedandchronichepatitisBpatientshavealsorevealedthatthefunctionalfate ofepitopespecificitiesdiffersmarkedlyinchronicinfection.Core18–27specificCD8 TcellscannotbedetectedinthecirculationwhenHBVDNAlevelsare>107copies/ ml.Amongstothers,weshowedinchapter4thattheinabilitytodetectcore18–27 specificCD8Tcells within the circulatory compartment is not due to preferential intrahepaticlocalization28.ThisstateofHBVspecificTcelltoleranceappearstobe regulated mainly by the quantity ofHBV replication present in chronic hepatitis B patients.TheimpactofviralloadonantiviralTcellresponseshasbeencharacterized inanimalmodelsofviralinfections(likeLCMV),whichshowthatsustainedpresence of viral antigens leads to a progressive functional decline of virusspecificCD8 responsesandultimatelyleadstovirusspecificTcelldeletion43.Similarly,inHBV infected patients, the frequency and function of circulating and intrahepaticHBV specificCD8TcellsisinverselyproportionaltothelevelofHBVDNA 28.HBeAg,a secretoryformofthenucleocapsidantigen,isproducedinlargeexcessduringHBV replication.ThetolerizingeffectofHBeAghasbeenwellcharacterizedinmice4445 and likely contributes to the low level of corespecificTcell responses present in HBeAg+chronicpatients. SincevaccinationwilllikelybetheleastexpensivewaytotreatchronicHBVinfection, currentlyalotofeffortsarebeingputinstudyingtheuseofnewlydevelopedDNA vaccinestostimulatetheimpairedTcellresponseinpatientswithchronicdisease. Dependingontheviralgenecarriersystem,DNA vaccinesfallintotwocategories. Viral(orbacterial)recombinantvaccinesaregeneticallyengineerednonpathogenic viruses modified by inserting a foreign viral gene, encoding the desired vaccine protein. In contrast, the naked DNA (plasmid) vaccines are vectors encoding viral antigens,whichbecomeexpressedintheimmunizedhost46.TheDNAvaccinesare

114 DISCUSSION

designedtoencodeviralproteinsthatareimportanttargetsforneutralisingantibodies andTcell responses after denovo syntheses of antigen inv ivo (fig 1). In rodent modelsforchronicHBVinfection,suchplasmidvaccinesencodingoneorseveral modifiedHBV envelope proteins have been shown to induce antibody production andCD4andCD8TcellresponsesthatwereabletoovercometolerancetoHBsAg, despite its relatively high presence in the serum 4749.However, recently after intramuscular immunization with a plasmid DNA encoding two envelope proteins, inpatientswithchronicHBVinfectiononlytransientHBVspecificTcellresponses wereobservedwithoutasustainedvirologiceffect50.Therefore,newapproachesare beinginvestigated.SomeofthemoresophisticatedDNAvaccinescombinesurface proteinswiththeimmunodominantcoreproteintobroadentheantigenicprofileand inducepotentHBVspecificCD4andCD8Tcellresponses 51.Also,codeliveryof cytokines with DNA vaccines may elicit the desired immune response. In animal models intradermal vaccination with a plasmid vaccine encoding aHBV surface antigenshowedenhancedTcellprimingwhenplasmidsencodingIFNγ,GMCSFor IL15werecodelivered5253.Othersfocusonreducingtheviralloadwithconventional antiviraltherapytoreducetheimpactofhighviralloadonTcellresponses,priorto administeringthevaccines54.Manyothervariables,likethemodeofimmunization, thescheduleofimmunization,andtheantigeniccompositionofthevaccineandits adjuvantsarecurrentlybeingevaluatedfortheirroleindesigningtheoptimalDNA vaccineforimmunotherapyinchronicHBVinfection. Alternatively, in chapters 6 and 7 we have attempted to boost the impairedTcell responses in patients with chronicHBV infection, using conventional antiviral therapy55.Therapeuticvaccinesusuallyrestricttheantigenicprofiletoafewepitopes thoughttobeessential.SinceimmuneresponsesinresolvedhepatitisBinfections areoftendirectedagainstseveralbutvariableepitopes36,thebestimmunestimulus maybethewholeviruswithaverybroadantigenicprofile.Ininvivoimmunization (IVI)ofCHBpatients,followingrapidandprofoundvirussuppressionbyinterferon lamivudine combination therapy, lamivudine was withdrawn intermittently during continued interferon therapy.The rationale being that profound virus suppression will reduce the antigenic pressure on the impairedTcell response, after which it mayeffectivelyrespondtothesudden,increasedantigenexpressionduringtheviral reboundwhenlamivudineiswithdrawn.AlthoughinitiallyIVIwasabletotransiently suppressviralreplicationintwopatientswithCHB(chapter6) 55,inasubsequent pilotstudythemagnitudeoftheinducedTcellresponsewasinsufficienttocausea sustainedvirologicaleffectinthemajorityofpatients(chapter7).However,sinceonly moderatevirussuppressionwasachieved,futureattemptstoreachimmunecontrol overthevirusmaybemoresuccessfulwhen,priortothelamivudineinterruptions,the HBVDNAlevelsaremoreprofoundlyreduced,andwhentheperiodsoflamivudine interruptionareshortenedtolimittheviralrebound.

115 CHAPTER9

Help B cell Antibody production CD4+ T cell

Help MHC II presentation

Antigenic CD40L / CD40 peptide Dendritic Cell MHC I presentation CTL response CD8+ T cell CD28 / B7 Processing ? TCR Cytokines TLR

CD 205 Antigenic peptide Polypeptide secretion Proteosyntheses

Myocyte DNA vaccine (plasmid)

DNA vaccine (plasmid)

Fig1.ThepossiblepathwaysofantigenpresentationfollowingDNAimmunization (modifiedfromRajcanietal46). Theantigensynthesizedinmyocytes(orepidermalcells)istransferredtoDCthat interactwithCD4TcellsviathemajorhistocompatibilityclassII(MHCII)molecule. TheseCD4TcellsprovidehelpatspecificprimingofCD8Tcellsand/orBcellsby meansofcytokinerelease.AntigenisbeingpresentedbyDCtoCD8Tcellsviathe MHCclassImolecule.DCcostimulateTcellsviatheB7andCD40receptorsthat interacts withCD28 andCD40L respectively, both present on the surface ofCD8 Tcells.The uptake of immunogenic peptides by DC is mediated by theCD 205 receptor.CytokineproductionbyDCisactivatedbymeansoftolllikereceptor(TLR) signalling.

Dendritic cells Dendriticcells(DC)representthemostpotentantigenpresentingcells,andthusplay animportantroleintheinductionofspecificTcellresponses56.Functionaldefects inDCcouldthereforebeanimportantmechanismofthevirustoevadehostimmune responses56andmayexplainthestateofTandBcellhyporesponsivenesspresent inchronichepatitisBpatients.Indeed,inchapter2wehaveshownthattwomajor DC precursors, the myeloid (mDC) and plasmacytoid (pDC) dendritic cells, are functionallyimpairedinpatientswithchronichepatitisB57.Thismightbeanimportant waybywhichHBVevadesanadequateimmuneresponseleadingtoviralpersistence

116 DISCUSSION

anddiseasechronicity.SinceproductiveHBVreplicationinDChasrecentlybeen excluded in chronic hepatitis B patients 58, further research should determine the mechanismbywhichHBVinterfereswiththeDCfunctions. After initial encouraging results using a mouse model for chronicHBV infection, recentlyadendriticcellbasedtherapeuticvaccinewasdevelopedforthetreatment ofpatientswithchronicdisease 59,60.Inthisapproachmonocytesareisolatedfrom the blood, then matured and pulsed withHBsAg invitro and finally administered tothepatients.ThisautologousDCvaccinewasabletoreducetheviralloadand induceHBeAgseroconversioninsomepatients 60.Nodataontheinductionofan HBVspecificTcellresponsewerereported.Therefore,manyfactorslikeantigenic compositionandthescheduleofimmunizationremaintobeoptimized,beforeDC vaccinationcanbeappliedasimmunotherapyforCHB.

Regulatory T cells Studiesofnumerousexperimentalmodelshaveprovidedevidencethatapopulation ofspecializedTcellsareabletoregulatetheimmuneresponse.Theseregulatory Tcells (Treg) reside mainly within a minor population ofCD4 cells that express the phenotypic markerCD25.They have been shown to suppress immunological responsesagainstself 61andforeignantigens 62throughsuppressivecytokinesor directcell–cellcontact;however,regulatoryeffectsofCD4+CD25+cellshavenot beenfullyelucidated.ItispossiblethatCD4+CD25+Tcellsareresponsibleforthe weakHBVspecificTcellresponseinchronichepatitisBpatientsandmayinhibitthe expansionandfunctionofHBVspecificCD8Tcells,precludingHBVclearancebut alsolimitingimmunemediatedliverdamage. TheimpactofcirculatingCD4+CD25+TcellsonHBVpathogenesishasrecently been analysed. Increased frequencies of circulating and intrahepatic regulatoryT cellsinpatientswithchronichepatitisBhavebeenshown 63 64.DepletionofCD4+ CD25+ cells increased the function ofHBVspecificTcells 63, 65.Combined, these datasuggestthataccumulationofTreginsidethelivermaycontributetoinhibitionof thelocalHBVspecificTcellresponse.Furthermore,itispossiblethatapopulation ofHBVspecific regulatory cells, different from theCD4+CD25+Tcell subset, analogous to the presence of IL10 producingHCVspecificTcells 66, might be inducedinchronicHBVinfection67. Inchapter8wehaveshownthatnonresponsetointerferonalphatherapymaybe relatedtotheinductionofTregduringtreatment.Thisopensupthepossibilitythat responseratestoIFNαmayimprovewhenTregfunctionissuppressed.Incancer, strategies that inhibit or depleteTregs and boost antitumour immunity are under investigation.Inmice,theremovalofCD4+CD25+TcellswithantiCD25depleting antibodies, in combination with antiCTLA4 antibodies, led to tumour rejection 68. However,acautionarynotecomesfromstudiesusingCTLA4blockadealongwith tumourspecificpeptidevaccinationsinhumanmelanomapatients,whichresultedin tumourregressioninsomepatientsbutalsosignificantdevelopmentofautoimmunity insixof14patients,thusemphasizingthedelicatebalancebetweentoleranceand

117 CHAPTER9

immunity 69.At present, no depleting antiCD25 antibodies are licensed for use in humans,buteventuallythesefindingsmayleadtonewimmunotherapeuticstrategies thatcanalsobeusedinthetreatmentofchronicHBVinfection.

Concluding remarks Even though virusspecificCD8Tcells play a major role inHBV clearance 12, coordinated activation of the different branches of adaptive immunity seems necessary to achieve viral control. When chronicity develops, diffuse defects of helperandcytotoxicTcellresponsesareapparentandarelikelytobemaintained bytheconcertedactionofhighlevelsofviralantigens,thepeculiarimmunological featuresoftheliverandbythecontributionofregulatorycellsordendriticcelldefects. FunctionalrestorationofthealteredHBVspecificimmunityduringchronicinfection, usingnewlydevelopedimmunotherapeuticstrategieswillthereforebeaverycomplex process.TherapeuticDNAvaccinationmayhavetobecombinedwithconventional therapy,cytokines,ortheuseofdendriticcellstoimprovethechancesofsuccess. Up to now all preclinical and clinical studies reported here, have been invariably verysafeandwelltolerated.Therehavebeennolifethreateningadverseevents, there has been no detectable organ pathology or systemic toxicity, and there has beennoautoimmunity,appearanceofantinuclearantibodiesorantibodiesagainst doublestrandedDNA 70.Furthermore,themostimportantriskofDNAvaccination, namelychromosomalintegrationandoncogenesishassofaronlybeentheoretical 70.However,itremainsofgreatimportancetocarefullyevaluatethelongtermeffects ofnewimmunotherapeuticstrategies,beforetheyareusedtoachievecontrolover chronicHBVinfection.

118 DISCUSSION

References 1. ChisariFV,FerrariC.HepatitisBvirusimmunopathogenesis.AnnuRevImmunol1995;13:2960. 2. BenhamouY,DohinE,LunelFabianiF,etal.EfficacyoflamivudineonreplicationofhepatitisBvirusin HIVinfectedpatients.Lancet1995;345:3967. 3. LaiCL,ChienRN,LeungNW,etal.AoneyeartrialoflamivudineforchronichepatitisB.AsiaHepatitis LamivudineStudyGroup.NEnglJMed1998;339:618. 4. AngusP,VaughanR,XiongS,etal.Resistancetoadefovirdipivoxiltherapyassociatedwiththeselection ofanovelmutationintheHBVpolymerase.Gastroenterology2003;125:2927. 5. van Zonneveld M,HonkoopP,Hansen BE, et al. Longterm followup of alphainterferon treatment of patientswithchronichepatitisB.Hepatology2004;39:80410. 6. JanssenHL,vanZonneveldM,SenturkH,etal.Pegylatedinterferonalfa2baloneorincombinationwith lamivudineforHBeAgpositivechronichepatitisB:arandomisedtrial.Lancet2005;365:1239. 7. AlexopoulouL,HoltAC,MedzhitovR,FlavellRA.RecognitionofdoublestrandedRNAandactivationof NFkappaBbyTolllikereceptor3.Nature2001;413:7328. 8. Lund J, SatoA,Akira S, MedzhitovR, IwasakiA.Tolllike receptor9mediated recognition ofHerpes simplexvirus2byplasmacytoiddendriticcells.JExpMed2003;198:51320. 9. Heil F,HemmiH,HochreinH, et al. Speciesspecific recognition of singlestrandedRNA via tolllike receptor7and8.Science2004;303:15269. 10. Moretta L, BottinoC,Pende D, Vitale M, Mingari MC, MorettaA.Human natural killer cells: Molecular mechanismscontrollingNKcellactivationandtumorcelllysis.ImmunolLett2005;100:713. 11. FongTL,DiBisceglieAM,BiswasR,etal.HighlevelsofviralreplicationduringacutehepatitisBinfection predictprogressiontochronicity.JMedVirol1994;43:1558. 12. ThimmeR,WielandS,SteigerC,etal.CD8(+)Tcellsmediateviralclearanceanddiseasepathogenesis duringacutehepatitisBvirusinfection.JVirol2003;77:6876. 13. GuidottiLG,RochfordR,ChungJ,ShapiroM,PurcellR,ChisariFV.Viralclearancewithoutdestructionof infectedcellsduringacuteHBVinfection.Science1999;284:8259. 14. BerquistKR,PetersonJM,MurphyBL,EbertJW,MaynardJE,PurcellRH.HepatitisBantigensinserum andliverofchimpanzeesacutelyinfectedwithhepatitisBvirus.InfectImmun1975;12:6025. 15. KajinoK,JilbertAR,SaputelliJ,AldrichCE,CullenJ,MasonWS.Woodchuckhepatitisvirusinfections: veryrapidrecoveryafteraprolongedviremiaandinfectionofvirtuallyeveryhepatocyte.JVirol1994;68: 5792803. 16. JilbertAR,WuTT,EnglandJM,etal.RapidresolutionofduckhepatitisBvirusinfectionsoccursafter massivehepatocellularinvolvement.JVirol1992;66:137788. 17. WielandS,ThimmeR,PurcellRH,ChisariFV.GenomicanalysisofthehostresponsetohepatitisBvirus infection.ProcNatlAcadSciUSA2004;101:666974. 18. WebsterGJ,ReignatS,MainiMK,etal.IncubationphaseofacutehepatitisBinman:dynamicofcellular immunemechanisms.Hepatology2000;32:111724. 19. CotePJ,ToshkovI,BellezzaC,etal.Temporalpathogenesisofexperimentalneonatalwoodchuckhepatitis virusinfection:increasedinitialviralloadanddecreasedseverityofacutehepatitisduringthedevelopment ofchronicviralinfection.Hepatology2000;32:80717. 20. NakamuraI,NuppJT,CowlenM,etal.Pathogenesisofexperimentalneonatalwoodchuckhepatitisvirus infection: chronicity as an outcome of infection is associated with a diminished acute hepatitis that is temporallydeficientfortheexpressionofinterferongammaandtumornecrosisfactoralphamessenger RNAs.Hepatology2001;33:43947. 21. MenneS,RonekerCA,RoggendorfM,GerinJL,CotePJ,TennantBC.Deficienciesintheacutephase cellmediatedimmuneresponsetoviralantigensareassociatedwithdevelopmentofchronicwoodchuck hepatitisvirusinfectionfollowingneonatalinoculation.JVirol2002;76:176980. 22. ChenH,PaulWE.CulturedNK1.1+CD4+TcellsproducelargeamountsofIL4andIFNgammaupon activationbyantiCD3orCD1.JImmunol1997;159:22409. 23. GonzalezAseguinolaza G, Van Kaer L, BergmannCC, et al. Natural killerTcell ligand alpha galactosylceramideenhancesprotectiveimmunityinducedbymalariavaccines.JExpMed2002;195: 61724. 24. Sharif S,Arreaza GA, ZuckerP, et al.Activation of natural killerTcells by alphagalactosylceramide treatmentpreventstheonsetandrecurrenceofautoimmuneType1diabetes.NatMed2001;7:105762. 25. GuidottiLG,ChisariFV.Tokillortocure:optionsinhostdefenseagainstviralinfection.CurrOpinImmunol 1996;8:47883. 26. GradyGF,LeeVA,PrinceAM,etal.HepatitisBimmuneglobulinforaccidentalexposuresamongmedical personnel:finalreportofamulticentercontrolledtrial.JInfectDis1978;138:62538. 27. Maini MK, BoniC, Ogg GS, et al. Direct ex vivo analysis of hepatitis B virusspecificCD8(+)Tcells

119 CHAPTER9

associatedwiththecontrolofinfection.Gastroenterology1999;117:138696. 28. WebsterGJ,ReignatS,BrownD,etal.LongitudinalanalysisofCD8+Tcellsspecificforstructuraland nonstructuralhepatitisBvirusproteinsinpatientswithchronichepatitisB:implicationsforimmunotherapy. JVirol2004;78:570719. 29. Kamal SM, Fehr J,Roesler B,PetersT,Rasenack JW.Peginterferon alone or with ribavirin enhances HCVspecificCD4Thelper1responsesinpatientswithchronichepatitisC.Gastroenterology2002;123: 107083. 30. MarinosG,TorreF,ChokshiS,etal.InductionofThelpercellresponsetohepatitisBcoreantigenin chronic hepatitis B: a major factor in activation of the host immune response to the hepatitis B virus. Hepatology1995;22:10409. 31. MichelML,ManciniBourgineM.TherapeuticvaccinationagainstchronichepatitisBvirusinfection.JClin Virol2005;34Suppl1:S10814. 32. Jung MC, Gruner N, ZachovalR, et al. Immunological monitoring during therapeutic vaccination as a prerequisite for the design of new effective therapies: induction of a vaccinespecificCD4+Tcell proliferativeresponseinchronichepatitisBcarriers.Vaccine2002;20:3598612. 33. ChisariFV,FerrariC.HepatitisBvirusimmunopathology.SpringerSeminImmunopathol1995;17:261 81. 34. BertolettiA, FerrariC, Fiaccadori F, et al.HLA class Irestricted human cytotoxicTcells recognize endogenouslysynthesizedhepatitisBvirusnucleocapsidantigen.ProcNatlAcadSciUSA1991;88: 104459. 35. NayersinaR,FowlerP,GuilhotS,etal.HLAA 2restrictedcytotoxicTlymphocyteresponsestomultiple hepatitisBsurfaceantigenepitopesduringhepatitisBvirusinfection.JImmunol1993;150:465971. 36. RehermannB,FowlerP,SidneyJ,etal.ThecytotoxicTlymphocyteresponsetomultiplehepatitisBvirus polymeraseepitopesduringandafteracuteviralhepatitis.JExpMed1995;181:104758. 37. HwangYK,KimNK,ParkJM,etal.HLAA21restrictedpeptidesfromtheHBxantigeninducespecificCTL responsesinvitroandinvivo.Vaccine2002;20:37707. 38. SprengersD,vanderMolenRG,KustersJG,etal.Flowcytometryoffineneedleaspirationbiopsies:a newmethodtomonitortheintrahepaticimmunologicalenvironmentinchronicviralhepatitis.JViralHepat 2005;12:50712. 39. SprengersD,MolenRG,KustersJG,etal.AnalysisofintrahepaticHBVspecificcytotoxicTcellsduring andafteracuteHBVinfectioninhumans.JHepatol2006. 40. MainiMK,BoniC,LeeCK,etal.TheroleofvirusspecificCD8(+)cellsinliverdamageandviralcontrol duringpersistenthepatitisBvirusinfection.JExpMed2000;191:126980. 41. KalamsSA,WalkerBD.ThecriticalneedforCD4helpinmaintainingeffectivecytotoxicTlymphocyte responses.JExpMed1998;188:2199204. 42. BertolettiA,SetteA,ChisariFV,etal.NaturalvariantsofcytotoxicepitopesareTcellreceptorantagonists forantiviralcytotoxicTcells.Nature1994;369:40710. 43. WherryEJ,BlattmanJN,MuraliKrishnaK,vanderMostR,AhmedR.ViralpersistencealtersCD8Tcell immunodominanceandtissuedistributionandresultsindistinctstagesoffunctionalimpairment.JVirol 2003;77:491127. 44. MilichDR,JonesJE,HughesJL,PriceJ,RaneyAK,McLachlanA.IsafunctionofthesecretedhepatitisB eantigentoinduceimmunologictoleranceinutero?ProcNatlAcadSciUSA1990;87:6599603. 45. ChenM,SallbergM,HughesJ,etal.ImmunetolerancesplitbetweenhepatitisBvirusprecoreandcore proteins.JVirol2005;79:301627. 46. Rajcani J, MoskoT,Rezuchova I.Current developments in viral DNA vaccines: shall they solve the unsolved?RevMedVirol2005;15:30325. 47. ManciniM,HadchouelM,DavisHL,WhalenRG,TiollaisP,MichelML.DNAmediatedimmunizationina transgenicmousemodelofthehepatitisBsurfaceantigenchroniccarrierstate.ProcNatlAcadSciUSA 1996;93:12496501. 48. Mancini M,Hadchouel M,TiollaisP, Michel ML.Regulation of hepatitis B virus mRNA expression in a hepatitis B surface antigen transgenic mouse model by IFNgammasecretingTcells after DNAbased immunization.JImmunol1998;161:556470. 49. LuM,HilkenG,KruppenbacherJ,etal.Immunizationofwoodchuckswithplasmidsexpressingwoodchuck hepatitisvirus(WHV)coreantigenandsurfaceantigensuppressesWHVinfection.JVirol1999;73:281 9. 50. ManciniBourgineM,FontaineH,ScottAlgaraD,PolS,BrechotC,MichelML.Inductionorexpansionof TcellresponsesbyahepatitisBDNAvaccineadministeredtochronicHBVcarriers.Hepatology2004;40: 87482. 51. ThermetA,RobaczewskaM,RollierC,etal.IdentificationofantigenicregionsofduckhepatitisBvirus

120 DISCUSSION

coreproteinwithantibodieselicitedbyDNAimmunizationandchronicinfection.JVirol2004;78:194553. 52. DuDW,JiaZS,LiGY,ZhouYY.HBVDNAvaccinewithadjuvantcytokinesinducedspecificimmune responsesagainstHBVinfection.WorldJGastroenterol2003;9:10811. 53. KwissaM,LindbladEB,SchirmbeckR,ReimannJ.CodeliveryofaDNAvaccineandaproteinvaccine withaluminumphosphatestimulatesapotentandmultivalentimmuneresponse.JMolMed2003;81: 50210. 54. HoriikeN,FazleAkbarSM,MichitakaK,etal.Invivoimmunizationbyvaccinetherapyfollowingvirus suppressionbylamivudine:anovelapproachfortreatingpatientswithchronichepatitisB.JClinVirol 2005;32:15661. 55. SprengersD,JanssenHL,KwekkeboomJ,NiestersHG,deManRA,SchalmSW.Invivoimmunization followingvirussuppression:anovelapproachforinducingimmunecontrolinchronichepatitisB.JViral Hepat2003;10:79. 56. BanchereauJ,BriereF,CauxC,etal.Immunobiologyofdendriticcells.AnnuRevImmunol2000;18:767 811. 57. vanderMolenRG,SprengersD,BindaRS,etal.Functionalimpairmentofmyeloidandplasmacytoid dendriticcellsofpatientswithchronichepatitisB.Hepatology2004;40:73846. 58. UntergasserA,ZedlerU,LangenkampA,etal.Dendriticcellstakeupviralantigensbutdonotsupport theearlystepsofhepatitisBvirusinfection.Hepatology2006;43:53947. 59. FazleAkbarSM,FurukawaS,OnjiM,etal.SafetyandefficacyofhepatitisBsurfaceantigenpulsed dendriticcellsinhumanvolunteers.HepatolRes2004;29:136141. 60. ChenM,LiYG,ZhangDZ,etal.Therapeuticeffectofautologousdendriticcellvaccineonpatientswith chronichepatitisB:aclinicalstudy.WorldJGastroenterol2005;11:18068. 61. SakaguchiS.RegulatoryTcells:keycontrollersofimmunologicselftolerance.Cell2000;101:4558. 62. SuvasS,KumaraguruU,PackCD,LeeS,RouseBT.CD4+CD25+Tcellsregulatevirusspecificprimary andmemoryCD8+Tcellresponses.JExpMed2003;198:889901. 63. StoopJN,vanderMolenRG,BaanCC,etal.RegulatoryTcellscontributetotheimpairedimmune responseinpatientswithchronichepatitisBvirusinfection.Hepatology2005;41:7718. 64. XuD,FuJ,JinL,etal.CirculatingandLiverResidentCD4+CD25+RegulatoryTCellsActivelyInfluence theAntiviralImmuneResponseandDiseaseProgressioninPatientswithHepatitisB.JImmunol2006; 177:73947. 65. FranzeseO,KennedyPT,GehringAJ,etal.ModulationoftheCD8+TcellresponsebyCD4+CD25+ regulatoryTcellsinpatientswithhepatitisBvirusinfection.JVirol2005;79:33228. 66. AccapezzatoD,FrancavillaV,ParoliM,etal.HepaticexpansionofavirusspecificregulatoryCD8(+)T cellpopulationinchronichepatitisCvirusinfection.JClinInvest2004;113:96372. 67. HyodoN,NakamuraI,ImawariM.HepatitisBcoreantigenstimulatesinterleukin10secretionbyboth TcellsandmonocytesfromperipheralbloodofpatientswithchronichepatitisBvirusinfection.ClinExp Immunol2004;135:4626. 68. SutmullerRP,vanDuivenvoordeLM,vanElsasA,etal.SynergismofcytotoxicTlymphocyteassociated antigen4blockadeanddepletionofCD25(+)regulatoryTcellsinantitumortherapyrevealsalternative pathwaysforsuppressionofautoreactivecytotoxicTlymphocyteresponses.JExpMed2001;194:823 32. 69. PhanGQ,YangJC,SherryRM,etal.CancerregressionandautoimmunityinducedbycytotoxicT lymphocyteassociatedantigen4blockadeinpatientswithmetastaticmelanoma.ProcNatlAcadSciUS A2003;100:83727. 70. HankeT.OnDNAvaccinesandprolongedexpressionofimmunogens.EurJImmunol2006;36:8069.

121

SUMMARY SAMENVATTING DANKWOORD CURRICULUMVITAE

SUMMARY

Summary

Despitethepresenceofanef fectivevaccinesince 1982,chronichepatitisBvirus infection(CHB)stillranksamongthehighestcausesofmortalityfrominfectious diseasesworldwide.Thestudiespresentedinthisthesiswereperformedtoget abetterinsightintotheantiviralimmuneresponseafterhepatitisBvirus(HBV) infection,andtoidentifyfactorsinthisimmuneresponsethatcontributetopersistent disease.Thesefactorsmayfunctionastargetsfornewimmunomodulatorytherapies thatimprovethelowresponseratestocurrentantiviraltreatmentoptions. Intheintroductiontothethesis(chapter 1)wehavedescribedthatavigorous, polyclonalandmultispecificcytotoxic(CTL)andhelperT(Th)cellresponsetoHBVis readilydetectableintheperipheralbloodofpatientswithacuteselflimitedhepatitis B,butisweak,antigenicallyrestrictedorundetectableinpatientswithchronicHBV infection.Wereviewedreporteddataonimmunotherapeuticstrategiestoimprove thisinefficientimmuneresponse,andconcludedthatatpresenttheonlyevident effectiveimmunomodulatorytherapyis(pegylated)interferonalpha(IFNα),which leadstosustainedresponseinonly3040%oftreatedpatients. Dendriticcells(DC)playanimportantroleintheinductionofantiviralimmune responses.Inchapter 2wecomparedthenumber,phenotypeandfunctionoftwo importantbloodprecursorDC,myeloidDC(mDC)andplasmacytoidDC(pDC),of chronichepatitisBpatientswithhealthyvolunteers.Nodifferencesinpercentagesof mDCandpDCinperipheralbloodmononuclearcellswereobservedbetweenchronic hepatitisBpatientsandhealthycontrols.However ,theallostimulatorycapacityof mDC,wassignificantlydecreasedinpatientscomparedtocontrols.Inaddition,mDC ofpatientsshowedareducedexpressionofcostimulatorymoleculesCD80and CD86,andreducedcapacitytoproducetumornecrosisfactorα.Purifi edpDCfrom patientsproducedlessinterferonα,animportantantiviralcytokine,thanpDCisolated fromcontrols.Therefore,mDCandpDCarefunctionallyimpairedinpatientswith chronichepatitisBandthismightbeanimportantmechanismbywhichHBVevades anadequateimmuneresponse,leadingtoviralpersistenceanddiseasechronicity. Informationaboutcharacterandgradeoftheintrahepaticimmuneresponsein viralhepatitisisimportantforevaluationofdiseasestageandef fectoftherapy . Complicationslikehaemorrhageprovidealimitationtofrequentlyperformingstandard tissueneedlebiopsies(TB),andhavemademanyinvestigatorsuseperipheral bloodassurrogatemarkerinstead,despitetheknowledgethatresidentintrahepatic populationsoflymphocytesdif fersignificantlyfrompopulationsinthecirculation. Fineneedleaspirationbiopsy(FN AB)hasprovedtobeaneasyandatraumatic methodforthediagnosisofacuterejectioninkidneyandlivertransplantrecipients, andpatientswithfocalliverlesions.Inchapter3weshowedthatfl owcytometry offineneedleaspirationbiopsyoftheliverallowsreliableanalysisoflymphocytes obtainedfromtheintrahepaticcompartment,inpatientswithviralhepatitis.Therefore, theFNABisavaluabletoolthatenablesustostudyinmoredetailtheimmunological eventsatthesiteofHBVreplication.

125

Baseduponbiochemicalandvirologicalparametersseveralpotentiallysuccessive phasesofchronicHBVhavebeenwelldescribed.Intheimmunetolerancephase serumhepatitisBsurfaceantigen(HBsAg)andhepatitisBeantigen(HBeAg)are detectable;serumHBV DNA levelsarehighandserumaminotransferasesare normalorminimallyelevated.Intheimmuneclearancephase,serumHBV DNA levelsdecreaseandserumaminotransferaselevelsincrease. At hirdphase,termed theinactivecarrierstateischaracterizedbylowlevelsofHBV DNA andnormal concentrationsofaminotransferases.Inchapter4weapplytheFNABprocedureto characterizethedifferentphasesofchronicHBVinfectionbyanalysingthecomposition ofkeypopulationsofimmuneeffectorcells,i.e.(HBV specific)CD8+Tcells,CD4+ TcellsandCD56+NaturalKiller(NK)cellsintheliver.Thefin dingssuggestthatthe clinicalphasesofchronicHBVinfection,havecharacteristicintrahepaticimmune responses.InimmunetolerantpatientsNKcellsarethedominantimmuneef fector cellpopulationintheliver,andintheimmuneclearancephasethepatientswithlow viralloadhadahigherproportionofintrahepaticCD4+Tcells,thanpatientswithhigh viralload.CD4+Tcellsmaythereforebeimportantinachievingimmunecontrolover thevirus. TheFNABprocedurewasalsousedinChapter5.Inthisstudyweaimedto characterizethevirusspecificCD8+Tcellresponseintheliverofpatientswithacute HBVinfection.DuringresolutionofacuteHBVinfectioninHLAA2positivepatients tetramerswereusedtoidentifyHBVspecificCD8+TcellsinFNABcytology.Atfir st presentationtherewasacorrelationbetweenthefrequencyofintrahepaticnon specificCD8+Tcellsandthedegreeofliverdamage.Duringandafterthedisease therewassequesteringofHBVspecificCD8+Tcellsintheliver,andthepercentage ofintrahepaticHLADRexpressingHBV specificCD8+ Tcellswashigherthanin PB.Three monthsafterHBsAgseroconversionthefrequencyofintrahepaticHBV specificCD8+ Tcellsremainedhigh. Togetherwiththefi ndinginchapter4,that lowfrequenciesofintrahepaticHBV specificCD8+Tcellswerealsofoundinsome immuneclearancepatientswithlowviralload,thesedatasuggestthatintrahepatic HBVspecificCD8+Tcellsplayanimportantroleinclearingthevirus. Inchapters6and7wehaveattemptedtoboosttheimpaired Tcellresponsesin patientswithchronicHBVinfection,usingconventionalantiviraltherapy .In invivo immunization(IVI)ofCHBpatients ,followingrapidvirussuppressionbyinterferon lamivudinecombinationtherapy,lamivudinewaswithdrawnintermittentlyduring continuedinterferontherapy .The rationalebeingthatprofoundvirussuppression willreducetheantigenicpressureontheimpaired Tcellresponse,afterwhichit mayeffectivelyrespondtothesudden,increasedantigenexpressionduringtheviral reboundwhenlamivudineiswithdrawn.Althoughinitially IVIwasabletotransiently suppressviralreplicationintwopatientswithCHB(chapter6),inasubsequent pilotstudythemagnitudeoftheinduced Tcellresponsewasinsufficienttocausea sustainedvirologicaleffectinthemajorityofpatients(chapter7). LittleisknownaboutwhytreatmentwithIFN αleadstoaresponseinonlyaminority ofpatientswithchronicHBV .ItwasrecentlyshownthatinthesepatientsCD4+

126 SUMMARY

CD25+regulatoryTcells(Treg)cansuppresstheHBV specificimmuneresponse. Inchapter8weaimedtoinvestigatewhetherinnonresponderstoIFNαtherapyTreg contributetotreatmentfailurebydownregulatingtheHBVspecificTcellresponses. FourteenpatientsreceivedpegylatedIFNαmonotherapyfor52weeksandwere followedupfor26weeks.Whereastherewasnodifferenceinthefrequenciesof CD4+CD25+Tregbetweenrespondersandnonrespondersatthestartoftreatment, innonrespondersandnotinresponderstherewasasignifi cantincreaseinthe frequencyofthesecellsduringtreatment.Incontrasttotheresponders,thenon respondersshowedasignificantincreaseinfrequencyofIL10producingcells.Treg depletionresultedinincreasedproliferationcapacityandincreasedfrequenciesof HBVspecificINFγproducingcells,butdidnotaffectthefrequencyofIL10producing cellsmeasuredduringthecourseofthetreatment.Thisstudyindicatesthatthere maybeanimportantroleforCD4+CD25+TregandIL10secretingregulatorycells, inHBVpersistenceduringandafterPegIFNαtherapy. Inchapter9wediscusstherecentinsightsintothepathogenesisofchronicHBV infection,reportedbyusandothers,andtheirpotentialasatargetforfuture immunotherapeuticstrategies.

127 Samenvatting

Ondankshetfeitdatersinds1982eeneffectiefvaccinbestaat,blijftchronische hepatitisBvirusinfectie(CHB)wereldwijdeenbelangrijkeoorzaakvoorsterfte.De studiesinditproefschriftwerdenuitgevoerdomeenbeterinzichtteverkrijgeninde antiviraleimmuunresponsnainfectiemethethepatitisBvirus(HBV),enomfactoren teidentificerendiebijdragenaanhetpersisterenvandeviraleleverziekte.Aangezien debeschikbareantiviralemedicijnenslechtsinbeperktemateef fectiefzijn,zouden dezefactorenalsdoelkunnendienenvoortoekomstigeantiviralemiddelenmeteen betertherapeutischeffect. Indeintroductievanditproefschrift(hoofdstuk1)wordtbeschrevendatde afweerreactieinhetbloedvanmensendieherstellenvaneenacuteHBVinfectie wordtgekenmerktdooreenhevige,policlonaleenmultispecifi ekecytotoxische T cel(CTL)enhelper Tcelrespons.Integenstellinghiermee,wordtbijpatiëntenmet CHBslechtseenbeperkte,ensomszelfshelemaalgeenTcelresponsaangetoond. Tevenswordterindithoofdstukgesprokenoverdeverschillendetherapeutische strategieënwaarmeeinhetverledenisgetrachtomdezezwakkeimmuunrespons testimuleren.Uitdegegevensblijktdatmomenteelinterferonalfa(IFNα)demeest effectieveimmuunmodulatoiretherapieis,meteensuccesvolleklaringvanhetvirus bij30a40%vandebehandeldepopulatie. Dendritischecellen(DC)zijnbelangrijkvoordeinductievandeantivirale afweerreactie.Inhoofdstuk2vergelijkenwedeaantallen,hetfenotypeendefunctie vantweetypenprecursorDC,myeloideDC(mDC)enplasmacytoideDC(pDC),uit hetbloedvanpatiëntenmetCHBmetdievangezondevrijwilligers.Depercentages mDCenpDCwarennietverschillend,maardeallostimulatoirecapaciteitvanmDC wassignificantverlaagdbijpatiëntentenopzichtevangezondevrijwilligers. Tevens bleekdatbijpatiëntendeexpressievandecostimulatoiremoleculenCD80enCD86 opmDCwasafgenomen,endatdezecellenmindergoedinstaatwarenomtumor necrosisfactoralfateproduceren.PlasmacytoideDCvanpatiëntenproduceerden minderinterferonalfa,eenbelangrijkeantiviralecytokine.Derhalveisbijpatiënten metchronischeHBVinfectiedefunctievanmDCenpDCafgenomen,enditzou kunnenbijdragenaandebeperkteafweerreactieenhetpersisterenvandeinfectiebij patiëntenmetchronischeHBV. Informatieoverdeaardvandeafweerreactieindeleverisbelangrijkvooreengoede evaluatievanhetstadiumvandeviraleleverziekteenheteffectvanbehandeling.Tot ophedenwerdfrequentediagnostiekvandeleverbeperktdoordecomplicatiesdie gepaardkunnengaanmethetuitvoerenvaneenstandaardleverbiopsie,waarbijvooral gedachtmoetwordenaanbloedingen.Veelonderzoekersmakendaaromgebruikvan cellenuithetbloed,ondanksdewetenschapdatdesamenstellingvanafweercellen indeleverenormverschiltvandieindecirculatie.Dedunnenaaldbiopsie(DNB)is eeneenvoudigeenweiniginvasieveprocedure,diealsindsenigetijdwordtgebruikt voorhetdiagnosticerenvanacuteafstotingnaeennierenlevertransplantatie,enbij patiëntenmetfocaleleverafwijkingen.Inhoofdstuk3wordtbesprokendatdeDNB

128 SAMENVATTING

eveneensgeschiktisombijpatiëntenmetviralehepatitisafweercellenuitdelever teisoleren,entonenweaandatdezecellenvervolgensbetrouwbaargeanalyseerd kunnenwordenmetbehulpvanflowcytometrie.Derhalvewordthetmogelijkomvia eenweiniginvasievemethodecelmateriaaluitdeleverteverkrijgenvooronderzoek naardeimmuunresponsopdeplaatsvanviralereplicatie. Opbasisvanbiochemischeenvirologischeparameterswordteronderscheid gemaakttussen3potentieelopeenvolgendefasenvanCHB.Indeimmuuntolerante fasezijnhethepatitisBsurfaceantigeen(HBsAg)enhethepatitisBeantigeen (HBeAg)detecteerbaarinhetserum,ishetserumniveauvanHBV DNAhoog,en zijndeaminotransferaseninhetserumvanpatiëntennormaalofslechtsminimaal verhoogd.IndeimmuunklaringsfaseneemthetniveauvanHBV DNAafenstijgt hetniveauvandeaminotransferaseninhetserum.Dederdefase,ookwelhet inactievedragerschapgenoemd,wordtgekenmerktdooreenlaagniveauvanhet HBVDNAennormalewaardenvandeserumaminotransferasen.Inhoofdstuk4 wordtdedunnenaaldbiopsiegebruiktombovengenoemdefasenvanHBVinfectie tekarakteriserenmetbetrekkingtotdesamenstellingvandeafweercellendiezich bevindenindelever .DaarbijwordtmetnamegekekennaardeverdelingvanCD4 positieve(CD4+)enCD8positieve(CD8+)TcellenenCD56positieveNaturalkiller (NK)cellen,aangezienvandezecelpopulatiesbekendisdatzeeenbelangrijkerol spelenindepathogenesevanCHBinfectie.Debevindingenindestudiesuggereren datdeverschillendefasenvandeaandoening,gepaardgaanmeteenkarakteristieke intrahepatischeimmuunrespons.IndeimmuuntolerantefasezijndeNKcellende dominantepopulatieafweercellenindelever .Patiëntenindeimmuunklaringsfase enmetlagevirustiters,blijkenvooraleenhogefrequentieaanCD4+ Tcelleninde levertehebbeninvergelijkingmetpatiëntenuitdezefaseenmeteenhoogHBV DNA.DeCD4+Tcellenzijnderhalvemogelijkbelangrijkvoorimmuuncontroleover hetvirus.LagefrequentiesHBV specifiekeCD8+ Tcellenwerdenaangetoondbij enkelepatiëntenmeteenlaagHBVDNA Inhoofdstuk5wordtdedunnenaaldbiopsietoegepastvooreengedetailleerde analysevandevirusspecifiekeCD8+Tcelresponsindelevervanpatiëntentijdens ennaeenacuteHBVinfectie.Ophetmomentvaneerstepresentatievandepatiënt indekliniekbleekdatereencorrelatieistussendematevanleverschade,gemeten aandehandvanserumaminotransferasen,endefrequentienonspecifi ekeCD8+ Tcellenindelever.GedurendedeinfectieenernawarenerhogefrequentiesHBV specifiekeCD8+Tcellenindelever,eneengrootdeelvandezecellenbrachtHLADR totexpressie,enwasdusgeactiveerd.ZelfsdriemaandennaHBsAgseroconversie bleefdefrequentieHBV specifiekeCD8+ Tcellenhoog.Dezebevindingen suggererendatHBVspecifiekeCD8+Tcellenindelevereenbelangrijkerolspelen bijdeklaringvanhetvirus. Indehoofdstukken6en7hebbenwegetrachtomdebeperkteafweerreactiebij patiëntenmetchronischeHBVinfectietestimuleren.Bijinvivoimmunization(IVI)van patiëntenmetCHBwordtdevirustiterverlaagdmeteencombinatiebehandelingvan lamivudineeninterferonalfa,waarnalamivudineintermitterendwordtonderbroken

129 tijdenscontinueinterferontherapie.Erwordtveronderstelddatonderdrukkingvan viralereplicatieresulteertineenafnamevandehoeveelheidviraleantigenenmet eenherstelvandegeremde Tcelresponstotgevolg.Nadataanvankelijkbijtwee patiënteneentijdelijkeimmuuncontroleoverhetviruswerdgeobserveerd(hoofdstuk 6),bleekineendaaropvolgendegroterestudiedatdegeïnduceerde Tcelrespons onvoldoendewasomeenlangdurigeffecttebewerkstelligen,bijdemeerderheidvan deonderzochtepatiënten(hoofdstuk7). HetisonbekendwaaromIFNαtherapieslechtsbijeenminderheidvandebehandelde patiëntenresulteertingenezing.RecentisaangetoonddatbijpatiëntenmetCHB CD4+CD25+regulatoireTcellen(Treg)instaatzijnomdeantiviraleimmuunrespons teremmen.Inhoofdstuk8vanditproefschriftwordtonderzochtofdezecellen bijdragenaanhetfalenvanIFNαtherapie.V eertienpatiëntenwerdengedurende 52wekenbehandeldmetgepegyleerdeIFNαmonotherapie52weken,waarnaze nogeens26wekenwerdengevolgd.V oorafgaandeaandebehandelingwarener geenverschillentussendemensendiegenazen(responders)endemensendieniet goedreageerdenopbehandeling(nonresponders)metbetrekkingtotdefrequenties CD4+CD25+ Treg.Echter,gedurendedetherapiebleekdefrequentie Treginhet bloedvannonresponderstoetenemen,terwijldezesterkafnambijderesponders. BovendienbleekdaterbijdenonresponderseentoenamewasvanhetaantalIL10 producerendeCD4+ Tcellen.Wanneerde Tregwerdenweggenomenbleekdater eentoenamewasvanHBV specifiekeproliferatieeninterferonγproductiedoor T cellen,terwijlhetaantalIL10producerendeCD4+Tcellenonveranderdbleef.Deze bevindingenindicerendatCD4+CD25+ TcellenenIL10producerende Tcellen mogelijkeenbelangrijkerolspelenbijhetpersisterenvanchronischeHBVinfectie tijdensbehandelingmetIFNα. Inhoofdstuk9bediscussiërenwedebevindingenvanbovengenoemdestudiesinde contextvangepubliceerdedatavanandereonderzoeksgroepen,enwebespreken depotentiëleaangrijpingspuntenvoortoekomstige,nieuweantiviralebehandelingen voorchronischeHBVinfectie.

130

DANKWOORD

Dankwoord

Ditproefschriftistotstandgekomenmetdesteunvanvelen,dieikopdezeplaatswil bedankenvoorhuninspanningen.

VooralwilikHarryJanssenbedankenvoorzijnvoortdurendegedrevenheidenzijn vermogenommijn,bijtijdenwijlen,onstuimigeenergieingoedebanenteleidenen tehouden.Harry,jegafmederuimteommijnideeenteverwoordenen,indienniet teexotisch,uittewerken,zonderdaarbijheteinddoeluithetoogteverliezen.Datgaf mijhetvertrouwenenhetenthousiasmeomindewijdewereldvanhetonderzoek mijnpersoonlijkegrenzenteverleggen,envandieervaringmaakikongetwijfeldook namijnpromotienogdankbaargebruik. Meerderemensenhebbenmijgedurendedepromotieinmeerofminderemate begeleid.NatuurlijkRenatevanderMolen…denkikaanmijntijdophetlab,dandenk ikaanjou.Hartstikkebedanktvoorjegezelligheid,binnenenbuitendewerkomgeving, jedaadkrachtenjegepassioneerdemaniervanmanagen.HansKusterswilik bedankenvoorzijnvertrouwenenderuimtedieikervaardeomte“experimenteren”, maarmetnameookvoordevlijmscherpe(ensomszelfspijnlijke)kritieken.Jaap KwekkeboomenLucvanderLaanwilikgraagbedankenvoordeleerzamediscussies endezeerbruikbareadviezen.ProfessorSchalmwasimmerinspirerendenzaldat vermoedelijkookaltijdblijven,zoalsderustendeweloverwogenraadvanRobde Manaltijdverhelderendzalzijn.Herenbedankt. Eenaanzienlijkdeelvandeinhetproefschriftbeschrevenwerkisuitgevoerdmet dehulpvanenkelemensendieikdaarvoorspeciaalwilbedanken,tewetenRekha Binda.PatrickBoor,PaulaBiesta,JeroenStoop,ShantaManchamenlastbutnot leastAliceKok.Damesenherenhetwaseengrootplezierommetjullietewerken. Degrotehoeveelheidpatiëntenmateriaalwerdgrotendeelsverzamelddoorde damesvan“hetresearchassistententeam”.IkwildaaromCockie,HeleenenAnneke graagbedankenvoordegezelligeenvruchtbaresamenwerking.Deonregelmatige ensomsverrassendepolibezoekenvandepatiëntenzoudennietingoedeordezijn afgerondzonderdeprettigeenfl exibelemedewerkingvandemensenopdepoli MDL.DaarvoorbenikEsther,Esther,Ronald,Minou,Ellen,Laksmie,Wilmaenmet nameNermindanookzeerdankbaar. Wanneereenofanderadministratiefprobleemmijweereenstotwanhoophad gedreven,danwarenMarionenMargrietvanhetsecretariaatHepatologiealtijd beschikbaarvooradviesenhulp.Daarommijndankvoorjulliebegrip,enspeciaal ookMarionbedanktvoorjeassistentiebijhetorganiserenvanditproefschrift. DeklinischestudieswerdenondermeerbegeleiddoordemensenvanhetClinicalTrial Bureau.MetnameElkewilikdaarombedankenvoordeprettigesamenwerking.

TerwijlhierinBostonhetregelmatigegeklettervanderegenenhetgalmenvande sirenesvandeonophoudelijkuitrukkendebrandweerwagensenambulancesmij doetwegdromen,denkikaannogzoveleanderendiemijnpromotietijdtoteen

133 onvergetelijkehebbengemaakt…zoalsLeon,metwieikgelukkignogsteedseen nimmereindigendgesprekvoeroveronzewerkzaamhedenenderestvanhetleven, Thjon,RudienDorine…dieonzekamerophetlabtoteenwaar“thuis”maakten, Arnoud…dieopgeheeleigenwijzekritiekafwisseldemetgezelligheid,Lindamet wiejegezelligkonbabbelen…ennatuurlijkPauline,opdewerkvloerontmoet,en sindsdienmijnallerliefsteexperiment. Kortom,hetwaseenergprettigewerksfeerwaarvooriknogveleanderemensen ookergdankbaarben,endieermedetoeheeftgeleiddatikmijn“promotieproces” voorspoedighebkunnenafronden.

DaveSprengers.

CURRICULUMVITAE

CURRICULUM VITAE

Deauteurvanditproefschriftwerdgeborenop1november1974teGoirle.Hijgroeide op inTilburg alwaar hij aan hetTheresiaLyceum hetVWO voltooide.Vervolgens vertrokhijnaarBelgiëomaanhetRijksuniversitairCentrumAntwerpengeneeskunde te studeren. Na het eerste kandidatuur verhuisde hij in1994 naar Nijmegen voor hetvervolgvandezestudieaandeKatholiekeUniversiteitNijmegen.Hetdoctoraal examen werd behaald in juli1998 en het arts examen in 2001 (cum laude). Gedurende de studie volgde hij een facultatieve klinische stage aan de afdeling GastrointestinaleChirurgievanhetPrinceofWalesHospitalinHongKongeneen klinischestageaandeafdelingGastroenterologievanhetGrooteSchuurHospitalte Kaapstad,ZuidAfrika.Vanaugustus1999totapril2000werdindeVerenigdeStaten eenonderzoeksstagegedaanaandeafdelingGastroenterologievandeUniversityof NorthCarolinateChapelHill,ondersupervisievanProf.R.B.Sartor.Hetonderwerp van dit project was: “de rol van de gastrointestinale microflora bij het ontstaan vancolitisinHLAB27transgeneratten”.Vanafapril2001werktehijalsassistent geneeskundigeinopleidingtotklinischonderzoeker(AGIKO)ondersupervisievan Prof.dr.H.L.A.Janssenaanhetonderzoekbeschreveninditproefschrift.Injanuari 2005werdbegonnenmetdeopleidingtotMaagDarmLeverartsinhetErasmusMC teRotterdam(opleiderProf.dr.E.J.Kuipers).

137