• Nem Talált Eredményt

and pathomechanism Psychological side effects of immune therapies:symptoms ScienceDirect

N/A
N/A
Protected

Academic year: 2022

Ossza meg "and pathomechanism Psychological side effects of immune therapies:symptoms ScienceDirect"

Copied!
7
0
0

Teljes szövegt

(1)

Psychological side effects of immune therapies:

symptoms and pathomechanism

David Kovacs

1,7

, Peter Kovacs

1,2,7

, Nora Eszlari

1,4

, Xenia Gonda

1,3

and Gabriella Juhasz

1,4,5,6

Immunotherapiesrevolutionisedthetreatmentofseveral disordersbutshowspecificside-effectprofileswhich frequentlyinvolvepsychologicalsymptoms.Longterm interferon-alpha(IFN-alpha)therapycancausewide-ranging psychiatricside-effectsfromfatigue,insomnia,anxietytofull- blowndepression.Thistreatment-emergentdepressionshares severalsymptomswithmajordepressivedisorder(MDD)witha predominanceofsomatic/neurovegetativesymptoms,andcan betreatedwithantidepressants.However,thisexperience directedresearchtoinflammatorymechanismsinMDD.MDD hasbeenconfirmedasaheterogeneousdisorderwitha subgroupofpatientssufferingfromlow-gradechronic inflammationandfrequentlyresistanttotraditional

antidepressanttreatment.Thusfutureresearchshoulddevelop strategiestoidentifythoseMDDpatientswhocouldbenefit fromdrugsactingthroughinflammatorypathways.

Addresses

1MTA-SENeuropsychopharmacologyandNeurochemistryResearch Group,HungarianAcademyofSciences,Budapest,Hungary

2NationalInstituteofOncology,Budapest,Hungary

3DepartmentofClinicalandTheoreticalMentalHealth,Ku´tvo¨lgyiClinical Center,SemmelweisUniversity,Ku´tvo¨lgyiu.4,Budapest,Hungary

4DepartmentofPharmacodynamics,SemmelweisUniversity,Budapest, Hungary

5NeuroscienceandPsychiatryUnit,UniversityManchester,Manchester, UK

6MTA-SE-NAPBGeneticBrainImagingMigraineResearchGroup, HungarianAcademyofSciences,SemmelweisUniversity,Budapest, Hungary

Correspondingauthor:Juhasz,Gabriella (Gabriella.Juhasz@manchester.ac.uk)

7Equallycontributed.

CurrentOpinioninPharmacology2016,29:97–103

ThisreviewcomesfromathemedissueonImmunomodulation EditedbyFulvioD’Acquisto

ForacompleteoverviewseetheIssueandtheEditorial Availableonline22ndJuly2016

http://dx.doi.org/10.1016/j.coph.2016.06.008

1471-4892/#2016TheAuthors.PublishedbyElsevierLtd.Thisisan openaccessarticleundertheCCBY-NC-NDlicense(http://creative- commons.org/licenses/by-nc-nd/4.0/).

Introduction

Immunotherapyisaspecialbiologicaltherapytargetedat activatingtheinnateimmunesystemtofightinfectionsor

cancer,ordownregulateimmuneresponseincaseofauto- immunedisordersorallergies.Immunotherapiesthatboost immuneresponseagainsttumourcellsorviruses arefre- quently associatedwithearlyneurovegetative symptoms characterised byfatigue, psychomotor slowing, anorexia andpain[1].Thesesymptomsshowasignificantoverlap with the manifestationsof the so-called sicknessbehav- iour,causedbytheactivationofproinflammatorycytokines duringinfectionsandincludingsymptomsoffatigue,an- hedonia, low mood, social isolation and irritability [2].

Sickness behaviour is considered an adaptive response to promotehealing byreducing energy expenditure to- wardsnotnecessaryactivitiesanddecreasingexploratory behaviour, thus it resembles a behavioural pattern very similar toanxiety anddepressive symptoms.Depressive componentsincludinganhedonia,heightenedpainsensi- tivity,andsocialavoidancearemeanttoconserveenergyto fight the infection,while theanxious components were developed to avoid further conflicts which might have negative outcome on thehealing process [3,4]. Thus, an evolutionary advantageous behavioural effect of im- muneresponse,whichenhancessurvival,isalsoadisturb- ingsideeffectoflifesaving immunotherapiesleadingto significantsuffering,burdenandlossofqualityoflife,and thus limiting the completion of the treatment course.

Becauseofthesharpincreaseinthenumberofdifferent immunotherapies and their indications in the present reviewwe focusonthepsychologicalsideeffectsof the mostfrequentlyinvestigatedinterferon-alpha(IFN-alpha) treatment andshortlysummarise the sideeffectsof the newlydevelopedimmunecheckpointblockingagents.

Proinflammatory cytokinesinthe therapy:

IFN-alpha

Interferons are a superfamily of proinflammatory cyto- kinesthatplayaroleinhostdefencemechanisms.IFN- alphaisanaturalcytokinewhichhasasyntheticversion:

IFN alpha-2b. IFN-alpha and IFN alpha-2b bind to interferon type-1receptors,activatingasignaltransduc- tionpathwayleadingtotheexpressionofmultiplegenes responsible for inhibition of tumour cell growth and proliferation [5]. IFN-alpha is widely usedin antiviral, forexample,hepatitisC[6],andantitumortherapiessuch as malignantmelanomaorhairycellleukaemia[7,8].

PsychologicalsymptomsduringIFN-alphatherapy Besidesearlyneurovegetativesymptomswhichmanifest inthemajorityofpatientsduringthefirstweeksofIFN- alphatreatmentas fatigue,painandanorexia,long-term

(2)

IFN-alphatreatmentoftencausesawidevarietyofpsy- chiatricside-effects,such as depression,fatigue,insom- nia, anxiety, and cognitive disturbances [1]. 10–40% of patients additionally develop afull depressive disorder syndromethatcanincludesuicidalideation,aboulia,lack ofmotivation,socialwithdrawal,guilt,anhedonia,irrita- bility,anxiety,andcrying[9].Mania,delirium,andpsy- chosisarefurtherbut lesscommonsideeffectsof IFN- alpha treatment. Approximately 30–70% of hepatitis C virus-infected patients treated with IFN-alpha experi- encedifferentdegreesofdepression.Mostofthemsuffer frommild ormoderate depressive symptoms, while se- veremajordepressionoccursinabout15% [10].

In addition to these similarities, the symptom profile (Table 1) oftreatment-emergentdepression andnatu- rally occurring major depressive episode show some distinctions[11].Namely,during long-termIFN-alpha treatmentpatientsreportedmoresevereweightlossand decreasedactivity,whilefeelingofguiltwaslesspromi- nentcomparedtomedicallyhealthydepressedsubjects [12].Thisobservationwassupportedbyarecentfinding whichsuggestedthatriskgeneticvariantintheIL-6gene morespecifically increaseddepressive symptomsmea- sured by the Zung Self-rating Depression Scale com- paredtotheBriefSymptomInventory,suggestingthat inflammatoryriskmechanismsare moreresponsiblefor somatic/neurovegetativesymptomsthancognitive-emo- tional signs of depression [13]. Furthermore, newly developedimmunecheckpointinhibitors,suchasanti- cytotoxicT-lymphocyteanti-gen4(CTLA-4)antibodies or humanised immunoglobulins against programmed death 1/ligand 1 (PD-1/PD-L1) which also enhance tumour-specific immune activity are associated witha

new category of side effects called ‘immune-related adverse events’ (irAE), in which the most frequent symptom is fatigue [14]. However, treatment-induced depressionhasnotbeendetectedinrelationtothesenew drugs[15].

Distressing and frequently untreated depression is a majorcontributortodosagereductionsortreatmentdis- continuationsduringIFN-alphatherapyandconsequent- lyincreasestheriskofineffectivetreatmentoutcomeor relapse[16,17].Thus,itisofgreatclinicalimportanceto investigate the mechanism underlying IFN-alpha-in- duceddepressionandpossiblepreventivestrategies.

PotentialmechanismsofIFN-alpha-treatment-induced depressivesymptoms

IFN-alpha is a strong activator of the proinflammatory cytokinesystembyincreasingtheperipheralconcentra- tionofinterlekin-6(IL-6),interleukin1-beta(IL-1b)and tumournecrosisfactor-alpha(TNF-a)[18].Recentneu- roimaging findingsshowed that acute administration of IFN-alpha elicited aninstant and profounddecrease in brainfunctional networkconnectivity whichresulted in changes in mood and cognitive symptoms [19]. In addition,long-termIFN-alphatreatment wasassociated withincreased glutamate levelin the basalganglia and dorsalanteriorcingulatecortex(dACC)[20]whichmight explainthepreviouslyreportedincreasedACCactivation and impaired error processing in IFN-alpha treated patients[21].

However,remainthequestionhowthecytokineimbal- anceinperipheralplasmasamples,inducedbyIFN-alpha treatment,couldspreadintothebrainwhichisprotected

Table1

Symptomprofileofsicknessbehaviour,majordepressivedisorder,IFN-alphainduceddepression,andpsychologicalsideeffectsof immunecheckpointinhibitors

Symptom domain Symptom Sickness

behaviour

MDD IFN-α ICI

Mood depressed mood x xxx xxx

anhedonia x xxx (x)

guilt x (x)

suicidal thoughts x (x)

Anxiety tension/irritability x x xx

fear x x xx

Cognitive memory/concentration x x

decision making x x

Somatic/neurovegetative appetite x x xxx

sleep xx x x

psychomotor retardation

xx x xxx

fatigue xx x xxx xxx

pain x x xxx

MDD: major depressive disorder, IFN-alpha: interferon alpha treatment, ICI: immune checkpoint inhibitor treatment, x: symptom is present, number of x: dominance of symptoms

MDD:majordepressivedisorder,IFN-alpha:interferonalphatreatment,ICI:immunecheckpointinhibitortreatment,x:symptomispresent,number ofx:dominanceofsymptoms.

(3)

by theblood–brain barrier(BBB) (Figure1).There are threemainproposedpathwayshowinflammatoryactiva- tioncanreachthebraintoexertitseffectonmood.First, cytokinemoleculescancrosstheBBBinsomeareasusing specifictransportproteins,andalsobynon-specifictrans- port in thecircumventricularorgans, including thearea postrema,orthesubfornicalorgan.Second,afferentnerve fibres(e.g.vagus)canalsocarrytheinflammatorysignals to thebrain wheninflammatorycytokinesbindto cyto- kinereceptorsandtransmitthesesignalsintothecentral nervous system [22].Third, there isa cellular pathway where whole activated immune cells can reach the brain withthehelp ofCC-chemokineligand 2(CCL2), and CXC-chemokine ligand 1 (CXCL1). Peripherial cytokines, such as TNF, can induce the transport by

activatingthemicroglialproductionofthesechemokines, andalsobyinflammatorystimulatedastrocytes[23].Post mortemstudiesofsuicidevictimswhoalsosufferedfrom depressionrevealedincreasedCCL2expressionandmac- rophage numbers in the perivascular space, suggesting increased transport of activated immune cells into the brain indepressivestate[24].

Decreasedneuralplasticity

BesidesactivatedcellscrossingtheBBB,microglialcells which have the original purpose of fighting infections inside theCNSalso produce cytokinessuchas TNF-a andIL-1b.Overactivationofmicroglialcellsiscommonly reportedinassociationwithdepressivestates.Prolonged elevation of cytokinelevelsinsidethebrain caninduce

Figure1

Social support

Monocyte

Plasticity ROS,RNS Neurotoxicity

Kynurenine IDO Microglia 5-HT

Humoral

Neural

Cellular

Teff Treg

NF-KB, BDNF Psychosocial

stress

Pathogen stress

Anti PD-1

Anti CTLA-4

BBB

Morphology AD

IL-1B IL-6 TNF

IFN-α IL-1B

TNF CCL2 CXCL1 IFN-α

therapy

Periphery Brain

Current Opinion in Pharmacology

Potentialroleofimmunemechanismsinimmunotherapy-inducedandstress-induceddepression.Activationoftheimmunesystemduringspecific immunotherapiesorbyinfectionsandsterilestressorslikechildhoodmaltreatment,recentnegativelifeeventsorchronicpaininducesdepression- relatedmechanisms.Theinflammatorysignalofactivatedimmunecellscancrosstheblood–brainbarrier(BBB),andreachthecentralnervous systembyvariouspathways.Themaincoordinatorsofthistransportarethemicroglialcells,producingattractingchemokines,andfacilitatingthe transportofactivatedimmunecellsthroughtheblood–brainbarrier.Microglialcellscanalsobeactivatedbypsychosocialstress.The

proinflammatorysignalinthebrainworksintheoppositedirectioncomparedtotheeffectofantidepressants,suppressing5-HTactivity,and activatingthekynureninepathway.Theneurotoxickynureninemetabolites,andthelowerBDNFandNF-KBlevelspromotereducedneural plasticity,andalsooxidativeradicalsdamageneuralpathwaysleadingtodepression-specificmorphologicalchanges.5-HT:serotonin,AD:

antidepressants,AntiCTLA-4:antibodyagainstanti-cytotoxicT-lymphocyteanti-gen4,AntiPD-1:antibodyagainstprogrammeddeath1/ligand1, BDNF:brainderivedneurotrophicfactors,CCL2:CC-chemokineligand2,CXCL1:CXC-chemokineligand1,IDO:indolamine-2,3-dioxygenase,IL- 1B:interleukin1-beta,IL-6:interlekin-6,INF-a:interferon-alpha,NF-KB:nuclearfactorkB,ROS:radicaloxygenspecies,RNS:radicalnitrogen species,Teff:effectorTlymphocytes,Treg:regulatoryTlymphocytes,TNF:tumournecrosisfactor.

(4)

neuralapoptoticpathwaysthroughtheireffectonnuclear factorkB(NF-kB)andbrain-derivedneurotrophicfactor (BDNF), resultingin impaired neuronalplasticity, and promoting depression-specific morphological alterations [25].

Decreasedneurotransmitteravailability

Proinflammatorycytokinescanalsoinfluenceneuralplas- ticitythroughtheactivationofindolamine-2,3-dioxygen- ase(IDO)whichistheratelimitingenzymeofkynurenine production.IDOisanenzymewhichconvertstryptophan intokynurenine,thuscompetingfortryptophanwiththe serotonine pathway [26]. Lower tryptophan availability itselfcancausedepressivesymptomsunderexperimental conditions,but lowerserotonergic function is also com- monlyfoundindepressedindividuals.

Increasedneurotoxicity

Besidestheloweramountoftryptophanleftforserotonin synthesis,activationofthe kynureninepathwayalsopro- duces potentially neurotoxic metabolites. For example, quinolinic acid which is elevated in post-mortem de- pressed suicide victims’ brain acts as an N-methyl-D- aspartate(NMDA) receptoragonist,andalsocontributes to disturbances in glutamate reuptake and release from astrocytes, leading to excitotoxic neuronal damage [27].

InterestingtonotethatIDOhyperactivitycorrelatedwith long-termdepression-specificsymptoms(depressedmood, anxiety) but not with early neurovegetative symptoms (pain,fatigue,anorexia)duringcytokinetherapy [28].

Increasedoxidativestress

Inflammatoryactivationisalsoaccompaniedbyincreased productionof radicaloxygen(ROS) andradical nitrogen species(RNS).Theactivityoftetrahydrobiopterin(BH4) whichisnecessaryformonoaminesynthesis,isreducedby oxidativeradicals,suggestinglowersynthesizingcapacity in the presence of inflammation. These oxidative sub- stancesaredamaging forDNA, fattyacids,andproteins alikeespeciallyinneuralcellswhicharethemostvulner- able to oxidative stress.Alterations in neuralcell mem- brane structure, serotonin binding capability, and intracellularsignallingmechanismsduetooxidativestress allcontributetoimpairedserotonergicactivityandtissue damage. Furthermore, superoxide anion radicals (SAR) behaveasaco-substrateofIDOcausingsuperinductionof the kynurenine pathway in the presence of excessive oxidative stress, thus oxidative radicals and kynurenine seemsto forma self-enhancingloop whichreduces the survivabilityofneuronalcellsgreatly[29].

DifferencebetweenthemodeofactionofIFN-alphaand immunecheckpointinhibitors

Althoughboth IFN-alphaand immune checkpoint inhi- bitors increase tumour specific immune response their psychological side-effect profiles are strikingly different.

IthasbeenrecentlydemonstratedthatCTLA-4antibodies

(e.g. ipilimumab) decrease the number of regulatory Tregcellsthroughnon-classicalmonocytes[30].Nonclassi- calorpatrollingmonocytesareresponsibleforclearingup the consequences of inflammation at the vascularendo- theliumandmaintainingtheintegrityoftheBBBthusthey might decrease expansion of the inflammation into the centralnervoussystem[31].PD-1/PD-L1antibodies(e.g.

nivolumab and pembrolizumab, both approved in 2014) increaseeffectorTcellactivitywithintissuesortumours wherecellsexpressPD-1/PD-L1whichtendstobelowin thebrain[14,32].

RiskfactorsofpsychologicalsideeffectsduringIFN- alphatreatment

Ithasbeenobservedthatbehaviouraleffectofinflamma- tion shows differences between patients. Thus knowl- edgeofrisk factorsfor developingdepressionwouldbe usefulinclinicalpracticetopreventsuchadverseeffects [33]. One major risk factor is presence of psychiatric disorders, especially depression in the medical history possibly because of shared biological mechanisms. For example,ithasbeendemonstratedthatincreasedpreva- lenceofdepressioninfemalesmightbeexplainedbythe greatersensitivityoffemalestoinflammatorysignalsboth atthebiochemicalandbehaviourallevel[34].

Potentialgeneticriskfactors

Geneticvariantsthathavebeenimplicatedindepression like theserotonin transportergene functional promoter polymorphism (5-HTTLPR) [35,36], or other serotonin (e.g. HTR1A) or inflammatory (e.g. IL-6, COX-2, TNF- alpha)pathwayrelatedgeneticvariantsincreasetherisk of developing psychological side effects during IFN- alpha treatment [33,37]. Furthermore, several genetic variantsthroughouttheinterferona/bsignallingpathway [38], and genetic variants in the IL-6, IL-1b or nitric oxidesynthase-1 (NOS1) genes increasedepressive and anxiety symptomatology, especially in the presence of psychosocialstressorsingeneralpopulations[13,39,40].

Psychosocialstress

Ithasbeenconsistentlyreportedthatpsychosocialstressis apotent inductorof inflammatoryresponse bothin ani- malsandhumans [41,42].Sterile stressors,like negative lifeevents,areabletoactivateinflammosomes,acomplex cytosolicproteincascadewiththeoriginalroleinpathogen host-defence reaction. The main products of inflammo- somes are inflammatory cytokines such as IL-1B, Il-6, TNF-a,andCRP[22,43].Thesecytokinesareconsidered tobeoneofthemoststablebiomarkersfordepression.In addition,stress exertsnotonly anacutebutalong-term effect on inflammation.For example, childhood trauma permanently upregulates proinflammatory cytokines, probablythroughepigeneticchanges[25,44].Inaddition, patientswithsensitizedstressresponsepathwaysaremore vulnerable to interferon IFN-alpha-induced depression.

For example, patients who responded to a first dose of

(5)

IFN-alphawith hyperactivityof thehypothalamic-pitui- tary-adrenalaxis(HPA)weresignificantlymorelikelyto developmajordepressionduringtreatmentthanpatients withmodeststresssystemresponsestotheinitialinjection [45].

Socialsupportasaprotectivefactor

It is important to note that social support is able to decreasetheriskofdepressionbothinageneralpopula- tion andinpatientstreatedwith immunotherapies[46].

Social supporthasbeen associatedwithdecreasedHPA activity and glucocorticoid concentration [47,48], with diminished dACC activityafter socialstressor [48], and withdecreasedproinflammatorycytokineproductiondur- ingstress[49].Indeed,melanomapatientswithincreased social support were less likely to develop depressive symptomsduring low-doseIFN-alphatreatment [50].

PreventionofIFN-alphainducedpsychological symptoms

Screeningandevaluationofriskfactorssuchaspsychiat- richistory,geneticpolymorphisms,premorbidelevations ininflammatorycytokines,orsocialsupportmayhelpto initiatepersonalizedpreventiontherapyinthosewhoare likely to develop psychiatric side effects during IFN- alphatreatment[9].Multiplestudiesfoundthatprophy- lactic andconcurrenttreatment withselectiveserotonin reuptake inhibitors (SSRIs) successfully reduced the incidence and severity of major depression in patients withchronichepatitisCinfectionormalignantmelanoma treatedwithIFN-alpha-2b[51].Inaddition,preliminary studiessuggestthatadietrichinomega-3polyunsaturat- edfattyacidswhichpromoteanti-inflammatoryprocesses inthebodyandmayalsoprocessmood-stabilisingeffects thusmaypreventsomaticdepressivesymptomsincyto- kine-treated patients [52]. However,it is interesting to note that (non IFN-alpha induced) major depressive patientswithanelevatedinflammatorybiomarkerprofile typicallyrespondpoortostandardantidepressantoranti- psychotictreatment [53].

Conclusionsofexperiencewithimmune-influencing agentsfortreatingmajordepressivedisorder

Introductionofimmunotherapieshavenotonlyadvanced thetreatment of tumours,virusinfections,autoimmune diseases andallergies but also shedlight on thepatho- mechanismofdepression.Nowitisincreasinglyaccepted thatmajordepressivedisorderisaheterogeneouscondi- tionconcerningbothitsmanifestationanditsunderpin- nings,andcoversseveraldistinctethiopathologicroutes, oneof thembeingchroniclowgrade-inflammation,that converge to the emergence of similar symptoms [4].

Indeed,SSRI-typeantidepressantsareabletocounteract theeffectofinflammatorymechanismsofdepressiontoa certain degree. For example there is experimental evi- dencethatreleaseofTNF-a,NO,andIL-6frommicro- glial cells in response to IFN-g administration can be

impaired bycertainSSRI-typeantidepressants[54],and they are able to shape the immune response in the peripheralimmunecellsaswell[55].However,asmen- tionedabove,anelevatedinflammatorybiomarkerprofile predictspoortreatmentresponsetofirstchoice(typically SSRI)antidepressants.Forexample,patientswithhigher C-reactiveproteinlevelsimprovedbetteronnortriptyline compared to escitalopram [56]. Thus, patients who are refractoryto standardantidepressant pharmacotherapies shouldbescreenedforinflammatorybiomarkerstoadjust their treatment. Supporting the alternative therapeutic approachtherearestudiesreportingcyclooxygenaseand nitric oxidesynthase inhibitorsexertingantidepressant- likeactivity[29].Inaddition,minocycline,anantibiotic substancewithcapabilityto reducemicroglialactivation wasabletorestorehippocampalneurogenesisandthusa potential candidatein depressiontreatment [57]. More- over, TNF-a antagonistinfliximab showed ahigherre- duction of depression scores compared to placebo in patients with treatment-resistantdepression and higher CRPlevels[58].Thusdrugstargetingtheinflammatory biologicalpathwaysmightbeawayforwardinasubsetof depressed patients who do not improve on standard antidepressanttreatment.

Conflictofinterest

David Kovacs is an employee of Gedeon Richter Plc.

MedicalDivision,butthecompanydidnotprovideany funding,orhadanyfurtherroleinthepreparationofthe article.Theotherauthorsdidnotdeclareanyconflicting interests.

Acknowledgements

ThisworkwassupportedbytheHungarianAcademyofSciences(MTA-SE NeuropsychopharmacologyandNeurochemistryResearchGroup);National DevelopmentAgency(KTIA_NAP_13-1-2013-0001)HungarianBrain ResearchProgramGrantNo.KTIA_13_NAP-A-II/14;andbythe HungarianAcademyofSciencesandtheHungarianBrainResearch ProgramGrantNo.KTIA_NAP_13-2-2015-0001(MTA-SE-NAPB GeneticBrainImagingMigraineResearchGroup).XeniaGondaisrecipient oftheJanosBolyaiResearchFellowshipoftheHungarianAcademyof Sciences.Thefundingbodiesfundedtheworkbuthadnoadditionalrolein thewritingofthisreview.

References and recommendedreading

Papersofparticularinterest,publishedwithintheperiodofreview, havebeenhighlightedas:

ofspecialinterest ofoutstandinginterest

1. CapuronL,GumnickJF,MusselmanDL,LawsonDH,

ReemsnyderA,NemeroffCB,MillerAH:Neurobehavioraleffects ofinterferon-alphaincancerpatients:phenomenologyand paroxetineresponsivenessofsymptomdimensions.

Neuropsychopharmacology2002,26:643-652.

2. DantzerR:Cytokine-inducedsicknessbehaviour:a

neuroimmuneresponsetoactivationofinnateimmunity.EurJ Pharmacol2004,500:399-411.

3. RaisonCL,MillerAH:Theevolutionarysignificanceof depressioninPathogenHostDefense(PATHOS-D).Mol Psychiatry2013,18:15-37.

(6)

4.

MillerAH,RaisonCL:Theroleofinflammationindepression:

fromevolutionaryimperativetomoderntreatmenttarget.Nat RevImmunol2015,16:22-34.

Thispaper demonstratesthatanevolutionaryadvantageousprocess, namelybehaviouralchangesduringinflammation,isanimportantpatho- mechanisminthedevelopmentofsocialstress-induceddepression.

5. SabelMS,SondakVK:Istherearoleforadjuvanthigh-dose interferon-alpha-2binthemanagementofmelanoma? Drugs 2003,63:1053-1058.

6. LinFC,YoungHA:,Interferons:Successinanti-viral

immunotherapy.CytokineGrowthFactorRev2014,25:369-376.

7. AlexandrescuDT,IchimTE,RiordanNH,MarincolaFM,Di NardoA,KabigtingFD,DasanuCA:Immunotherapyfor melanoma:currentstatusandperspectives.JImmunother 2010,33:570-590.

8. PestkaS:Theinterferons:50yearsaftertheirdiscovery,there ismuchmoretolearn.JBiolChem2007,282:20047-20051.

9. LotrichFE:Psychiatricclearanceforpatientsstartedon interferon-alpha-basedtherapies.AmJPsychiatry2013, 170:592-597.

10. SchaeferM,CapuronL,FriebeA,Diez-QuevedoC,RobaeysG, NeriS,FosterGR,KautzA,FortonD,ParianteCM:HepatitisC infection,antiviraltreatmentandmentalhealth:aEuropean expertconsensusstatement.JHepatol2012,57:1379-1390.

11. NorciniPalaA,StecaP,BagrodiaR,HelpmanL,ColangeliV, VialeP,WainbergML:Subtypesofdepressivesymptomsand inflammatorybiomarkers:anexploratorystudyonasampleof HIV-positivepatients.BrainBehavImmun2016.

12. CapuronL,FornwaltFB,KnightBT,HarveyPD,NinanPT, MillerAH:Doescytokine-induceddepressiondifferfrom idiopathicmajordepressioninmedicallyhealthyindividuals?

JAffectDisord2009,119:181-185.

13. KovacsD,EszlariN,PetschnerP,PapD,VasS,KovacsP, GondaX,BagdyG,JuhaszG:Interleukin-6promoter polymorphisminteractswithpainandlifestressinfluencing depressionphenotypes.JNeuralTransm(Vienna)2016, 123:541-548.

14. PostowMA,CallahanMK,WolchokJD:Immunecheckpoint blockadeincancertherapy.JClinOncol2015,33:1974-1982.

15. KovacsP,PanczelG,BorbolaK,JuhaszG,LiszkayG:

Psychologicalchangesinmelanomapatientsduring ipilimumabtreatmentcomparedtolow-doseinterferonalpha therapyafollow-upstudyoffirstexperiences.PatholOncol Res2014,20:939-944.

16. BaraldiS,HepgulN,MondelliV,ParianteCM:Symptomatic treatmentofinterferon-alpha-induceddepressioninhepatitis C:asystematicreview.JClinPsychopharmacol2012,32:531-543.

17. GarbeC,RadnyP,LinseR,DummerR,GutzmerR,UlrichJ, StadlerR,WeichenthalM,EigentlerT,EllwangerUetal.:Adjuvant low-doseinterferon{alpha}2awithorwithoutdacarbazine comparedwithsurgeryalone:aprospective-randomized phaseIIIDeCOGtrialinmelanomapatientswithregional lymphnodemetastasis.AnnOncol2008,19:1195-1201.

18. RaisonCL,CapuronL,MillerAH:Cytokinessingtheblues:

inflammationandthepathogenesisofdepression.Trends Immunol2006,27:24-31.

19.

DipasqualeO,CooperEA,TibbleJ,VoonV,BaglioF,BaselliG, CercignaniM,HarrisonNA:Interferon-alphaacutelyimpairs whole-brainfunctionalconnectivitynetworkarchitecture a preliminarystudy.BrainBehavImmun2015.

AnelegantpharmacologicalbrainMRIstudydemonstratedthatthefirst doseofIFN-alphainhepatitisCpatientscan disconnectfunctionally coupledbrainregionswithinhours.

20. HaroonE,WoolwineBJ,ChenX,PaceTW,ParekhS,SpiveyJR, HuXP,MillerAH:IFN-alpha-inducedcorticalandsubcortical glutamatechangesassessedbymagneticresonance spectroscopy.Neuropsychopharmacology2014,39:1777-1785.

21. CapuronL,PagnoniG,DemetrashviliM,WoolwineBJ, NemeroffCB,BernsGS,MillerAH:Anteriorcingulateactivation

anderrorprocessingduringinterferon-alphatreatment.Biol Psychiatry2005,58:190-196.

22. MillerAH,MaleticV,RaisonCL:Inflammationandits discontents:theroleofcytokinesinthepathophysiologyof majordepression.BiolPsychiatry2009,65:732-741.

23. D’MelloC,LeT,SwainMG:Cerebralmicrogliarecruit monocytesintothebraininresponsetotumornecrosis factoralphasignalingduringperipheralorganinflammation.J Neurosci2009,29:2089-2102.

24. Torres-PlatasSG,CruceanuC,ChenGG,TureckiG,MechawarN:

Evidenceforincreasedmicroglialprimingandmacrophage recruitmentinthedorsalanteriorcingulatewhitematterof depressedsuicides.BrainBehavImmun2014,42:50-59.

25. CattaneoA,MacchiF,PlazzottaG,VeronicaB,Bocchio- ChiavettoL,RivaMA,ParianteCM:Inflammationandneuronal plasticity:alinkbetweenchildhoodtraumaanddepression pathogenesis.FrontCellNeurosci2015,9:40.

26. VecseiL,SzalardyL,FulopF,ToldiJ:KynureninesintheCNS:

recentadvancesandnewquestions.NatRevDrugDiscov2013, 12:64-82.

27. TavaresRG,TascaCI,SantosCE,AlvesLB,PorciunculaLO, EmanuelliT,SouzaDO:Quinolinicacidstimulates synaptosomalglutamatereleaseandinhibitsglutamate uptakeintoastrocytes.NeurochemInt2002,40:621-627.

28. RaisonCL,DemetrashviliM,CapuronL,MillerAH:

Neuropsychiatricadverseeffectsofinterferon-alpha:

recognitionandmanagement.CNSDrugs2005,19:105-123.

29. LeonardB,MaesM:Mechanisticexplanationshowcell- mediatedimmuneactivation,inflammationandoxidativeand nitrosativestresspathwaysandtheirsequelsand

concomitantsplayaroleinthepathophysiologyofunipolar depression.NeurosciBiobehavRev2012,36:764-785.

30. RomanoE,Kusio-KobialkaM,FoukasPG,BaumgaertnerP, MeyerC,BallabeniP,MichielinO,WeideB,RomeroP,SpeiserDE:

Ipilimumab-dependentcell-mediatedcytotoxicityof regulatoryTcellsexvivobynonclassicalmonocytesin melanomapatients.ProcNatlAcadSciUSA2015, 112:6140-6145.

31. ThomasG,TackeR,HedrickCC,HannaRN:Nonclassical patrollingmonocytefunctioninthevasculature.Arterioscler ThrombVascBiol2015,35:1306-1316.

32. SunshineJ,TaubeJM:PD-1/PD-L1inhibitors.CurrOpin Pharmacol2015,23:32-38.

33. SmithKJ,NorrisS,O’FarrellyC,O’MaraSM:Riskfactorsforthe developmentofdepressioninpatientswithhepatitisCtaking interferon-alpha.NeuropsychiatrDisTreat2011,7:275-292.

34. MoieniM,IrwinMR,JevticI,OlmsteadR,BreenEC, EisenbergerNI:Sexdifferencesindepressiveand socioemotionalresponsestoaninflammatorychallenge:

implicationsforsexdifferencesindepression.

Neuropsychopharmacology2015,40:1709-1716.

35. JuhaszG,GondaX,HullamG,EszlariN,KovacsD,LazaryJ, PapD,PetschnerP,ElliottR,DeakinJFetal.:Variabilityinthe effectof5-HTTLPRondepressioninalargeEuropean population:theroleofage,symptomprofile,typeandintensity oflifestressors.PLoSOne2015,10e0116316.

36. LotrichFE,FerrellRE,RabinovitzM,PollockBG:Riskfor depressionduringinterferon-alphatreatmentisaffectedby theserotonintransporterpolymorphism.BiolPsychiatry2009, 65:344-348.

37. UdinaM,Moreno-EspanaJ,NavinesR,GimenezD,LangohrK, GratacosM,CapuronL,delaTorreR,SolaR,Martin-SantosR:

Serotoninandinterleukin-6:theroleofgenetic

polymorphismsinIFN-inducedneuropsychiatricsymptoms.

Psychoneuroendocrinology2013,38:1803-1813.

38.

MostafaviS,BattleA,ZhuX,PotashJB,WeissmanMM,ShiJ, BeckmanK,HaudenschildC,McCormickC,MeiRetal.:TypeI interferonsignalinggenesinrecurrentmajordepression:

(7)

increasedexpressiondetectedbywhole-bloodRNA sequencing.MolPsychiatry2014,19:1267-1274.

Thisstudyusedwholegenomeexpressiondataandpathwayanalysis methodstoshowthatinterferona/bsignallingpathwayisupregulatedin recurrentMDDpatientscomparedtocontrols.

39.

KovacsD,EszlariN,PetschnerP,PapD,VasS,KovacsP, GondaX,JuhaszG,BagdyG:EffectsofIL1Bsinglenucleotide polymorphismsondepressiveandanxietysymptomsare determinedbyseverityandtypeoflifestress.BrainBehav Immun2016,56:96-104.

A further supportthat genetic variants intheinflammatory pathway, namelypolymorphismsintheIL1Bgene,increasetheriskofdepressive andanxietysymptomsinthefaceofstressfullifeevents.

40. SarginsonJE,DeakinJF,AndersonIM,DowneyD,ThomasE, ElliottR,JuhaszG:Neuronalnitricoxidesynthase(NOS1) polymorphismsinteractwithfinancialhardshiptoaffect depressionrisk.Neuropsychopharmacology2014, 39:2857-2866.

41. DeinzerR,GranrathN,StuhlH,TworkL,IdelH,WaschulB, HerforthA:AcutestresseffectsonlocalIl-1betaresponsesto pathogensinahumaninvivomodel.BrainBehavImmun2004, 18:458-467.

42. MaesM,SongC,LinA,DeJonghR,VanGastelA,KenisG, BosmansE,DeMeesterI,BenoyI,NeelsHetal.:Theeffectsof psychologicalstressonhumans:increasedproductionofpro- inflammatorycytokinesandaTh1-likeresponseinstress- inducedanxiety.Cytokine1998,10:313-318.

43. IwataM,OtaKT,LiXY,SakaueF,LiN,DutheilS,BanasrM, DuricV,YamanashiT,KanekoKetal.:Psychologicalstress activatestheinflammasomeviareleaseofadenosine triphosphateandstimulationofthepurinergictype2X7 receptor.BiolPsychiatry2016,80:12-22.

44. DaneseA,MoffittTE,ParianteCM,AmblerA,PoultonR,CaspiA:

Elevatedinflammationlevelsindepressedadultswitha historyofchildhoodmaltreatment.ArchGenPsychiatry2008, 65:409-415.

45. CapuronL,RaisonCL,MusselmanDL,LawsonDH,NemeroffCB, MillerAH:AssociationofexaggeratedHPAaxisresponseto theinitialinjectionofinterferon-alphawithdevelopmentof depressionduringinterferon-alphatherapy.AmJPsychiatry 2003,160:1342-1345.

46. CapuronL,RavaudA,MillerAH,DantzerR:Baselinemoodand psychosocialcharacteristicsofpatientsdeveloping depressivesymptomsduringinterleukin-2and/orinterferon- alphacancertherapy.BrainBehavImmun2004,18:205-213.

47. GiesbrechtGF,PooleJC,LetourneauN,CampbellT,KaplanBJ, TeamAPS:Thebufferingeffectofsocialsupporton hypothalamic-pituitary-adrenalaxisfunctionduring pregnancy.PsychosomMed2013,75:856-862.

48. EisenbergerNI,TaylorSE,GableSL,HilmertCJ,LiebermanMD:

Neuralpathwayslinksocialsupporttoattenuated neuroendocrinestressresponses.Neuroimage2007, 35:1601-1612.

49. MoieniM,IrwinMR,JevticI,BreenEC,ChoHJ,ArevaloJM,MaJ, ColeSW,EisenbergerNI:Traitsensitivitytosocial

disconnectionenhancespro-inflammatoryresponsestoa randomizedcontrolledtrialofendotoxin.

Psychoneuroendocrinology2015,62:336-342.

50.

KovacsP,PanczelG,BalatoniT,LiszkayG,GondaX,BagdyG, JuhaszG:Socialsupportdecreasesdepressogeniceffectof low-doseinterferonalphatreatmentinmelanomapatients.J PsychosomRes2015,78:579-584.

Socialsupportasapositiveenvironmentaleffectisabletomoderatethe depressogeniceffectofactivationinthepro-inflammatorycytokinepath- way,possiblybyactingthroughoverlappingbiologicalprocesses.

51.

SarkarS,SchaeferM:Antidepressantpretreatmentforthe preventionofinterferonalfa-associateddepression:a systematicreviewandmeta-analysis.Psychosomatics2014, 55:221-234.

Thisreviewdemonstratedthatpreventiveantidepressanttreatmentdur- ingIFN-alphatherapyeffectivelypreventedIFN-associateddepressionin allpatientsirrespectiveofpre-existingpsychiatricsymptoms,thuscanbe offeredtoallpatients.

52. ChangJP,LaiHC,YangHT,SuWP,PengCY,GaleckiP, WalczewskaA,ParianteCM,SuKP:Polyunsaturatedfattyacids levelsandinitialpresentationofsomaticsymptomsinduced byinterferon-alphatherapyinpatientswithchronichepatitisC viralinfection.NutrNeurosci2015.

53. ParianteCM:Neuroscience,mentalhealthandtheimmune system:overcomingthebrain-mind-bodytrichotomy.

EpidemiolPsychiatrSci2015:1-5.

54. LeonardBE:Impactofinflammationonneurotransmitter changesinmajordepression:aninsightintotheactionof antidepressants.ProgNeuropsychopharmacolBiolPsychiatry 2014,48:261-267.

55.

DiRossoME,PalumboML,GenaroAM:Immunomodulatory effectsoffluoxetine:anewpotentialpharmacologicalaction foraclassicantidepressantdrug? PharmacolRes2016, 109:101-107.

This reviewemphasises that theantidepressant fluoxetineeffectively modulatesthefunctionofperipheralimmunecellsandmayhavearole inantitumorT-cellactivity.

56. UherR,TanseyKE,DewT,MaierW,MorsO,HauserJ, DernovsekMZ,HenigsbergN,SoueryD,FarmerAetal.:An inflammatorybiomarkerasadifferentialpredictorofoutcome ofdepressiontreatmentwithescitalopramandnortriptyline.

AmJPsychiatry2014,171:1278-1286.

57. HusainMI,ChaudhryIB,RahmanRR,HamiraniMM,QurashiI, KhosoAB,DeakinJF,HusainN,YoungAH:Minocyclineasan adjunctfortreatment-resistantdepressivesymptoms:study protocolforapilotrandomisedcontrolledtrial.Trials2015, 16:410.

58. RaisonCL,RutherfordRE,WoolwineBJ,ShuoC,SchettlerP, DrakeDF,HaroonE,MillerAH:Arandomizedcontrolledtrialof thetumornecrosisfactorantagonistinfliximabfortreatment- resistantdepression:theroleofbaselineinflammatory biomarkers.JAMAPsychiatry2013,70:31-41.

Ábra

Figure 1 Social support Monocyte Plasticity ROS,RNS NeurotoxicityKynurenineIDOMicroglia5-HTHumoralNeuralCellularTeffTregNF-KB,BDNFPsychosocialstressPathogenstress Anti PD-1 Anti CTLA-4 BBB MorphologyADIL-1BIL-6TNFIFN-αIL-1BTNFCCL2CXCL1IFN-αtherapyPeriphery

Hivatkozások

KAPCSOLÓDÓ DOKUMENTUMOK

Psychopharmacology (Berl). Youngstedt, S.D., Effects of exercise on sleep. Mendlewicz, J., Sleep disturbances: core symptoms of major depressive disorder rather than associated or

Since we identified regulation of interferon (IFN) response genes as one of the most significant pathways associated with ED treatment (Fig. 1c and Additional file 6: Table S4),

We report implementation of stereotactic body radiother- apy (SBRT) for the treatment of early, localized prostate cancer patients, and acute side effects caused by radiation

3.3.1.1 Analyses of the examination of muscle dysmorphia symptoms, eating disorder variables, and body attitudes

The purpose of the second study was to investigate the correlation of postpartum depressive and anxiety symptoms with maternal perception of the infant and the

Circadian Locomotor Output Cycles Kaput (CLOCK) Since depression is often characterized by a distur- bance of the circadian rhythms and sleep deprivation and bright light therapy

The second study was the first to demonstrate that ipilimumab elicited fewer psychological side-effects compared to interferon-alpha immunotherapy which suggests a better

To further investigate how social support modulates the depressogenic side effect of interferon treatment two social support groups were classified according to