60
8 th International forum for RA (IFRA) Welcome to an international meeting focused on Rheumatoid Arthritis Saturday September 23 d Tuesday September 26 th 2017 Aula Medica, Karolinska Institutet, Stockholm, Sweden www.RAforum2017.com Official program Organizers: Rheumatology Karolinska (main responsible prof Lars Klareskog) together with co- organizers prof Zhanguo Li, (Beijing) and prof Kazuhiko Yamamoto (Tokyo)

8 International forum for RA (IFRA) Rheumatoid …h24-files.s3.amazonaws.com/240496/991074-xMREL.pdf8th International forum for RA (IFRA) Welcome to an international meeting focused

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

8th International forum for RA

(IFRA)

Welcome to an international meeting

focused on

Rheumatoid Arthritis

Saturday September 23d – Tuesday September 26th 2017

Aula Medica, Karolinska Institutet, Stockholm, Sweden

www.RAforum2017.com

Official program

Organizers: Rheumatology Karolinska (main responsible prof Lars Klareskog) together with co-

organizers prof Zhanguo Li, (Beijing) and prof Kazuhiko Yamamoto (Tokyo)

www.RAforum2017.com

Most welcome to the 8th International Forum for Rheumatoid Arthritis, this time in Stockholm. The meeting aims to present progress in RA research and outline prospects and plans for coming years. A major focus of the meeting is to present and discuss research on the causes of the dis-ease, and of the gradual development of various symptoms with the hope that such knowledge will not only help to treat the disease earlier and better, but ultimately also prevent its occur-rence. Meetings with the name RA Forum have since 2009 been organized in China (Beijing) and in 2016 in Japan (Tokyo) and the present meeting in Stockholm aims to further de-velop the scope of RA Forum, by assembling clinicians and scientists from all over the world with a special interest in the disease RA – which so far has rarely been the entire focus of internation-al meetings. We believe that this focus will allow us to make and create new contacts, visions and joint pro-jects in RA research. The meeting is organized in several sessions reaching from epidemiology to molecular sciences and includes also sessions on treatment and prevention. As a major purpose of the meeting is to discuss and define new research agenda, we have reserved time in the end of each session for dis-cussion on the presented results and implication for further research – all led by the chairper-sons of the session. We hope that this somewhat different organization of the meeting with a big role of the chairper-sons, will enable many forward-looking discussions and allow us to leave the meeting with new ideas on how to further develop research care and prevention of RA. Yu are all most welcome !! Lars Klareskog, on behalf of the rest of the organising committee With financial support from

 

 

www.RAforum2017.com

8th International Forum for RA 2017 - Program

12:30

13:00–14:00

Registrationopen

Lunch

Saturday, September 23, 2017 

14:00–14:15 WelcomeandintroductiontothehistoryofandpurposeofIFRA

LarsKlareskog,ZhanguoLi,KazuhikoYamamoto

JIMWelcomeaddress

AndersEkbom,ProfessorKI,MemberoftheeditorialboardofJournalofInternalMedicine

Session 1: The good aspects of the situation today and how we arrived here 

14:15–14:45

Chairs:ZhanguoLi,KazuhikoYamamoto

Fromideasinresearchtoclinicalbenefits.Thelongtermperspectives

SirRavinderMaini,KennedyInstitute,UniversityofOxford

Session 2: The unresolved clinical and scientific problems of today – and why are they unresolved? 

Chairs:ToreKvien,OsloUniversityandAlisonKent,PARE/EULAR

14:50–15:15Patients’needsandcurrentscientificapproachestoRA;Notalwaysaligned.

JohanAskling,KarolinskaInstitutet,Stockholm

15:15–15:40Unresolvedproblemsasseenfromtheclinician’sperspective

MereteHetland,CopenhagenUniversity

15:40–16:05Scientificunresolvedproblemsinscience

ChrisBuckley,UniversityofBirmingham

16:05–16:30

Discussionsonneedfornovelsolutionstoadessthemostimportantunresolvedquestions

Discussionledbychairpersons

16:30–17.00 Afternoonbreakwithteaandcoffee

Session 3: Why do we get RA (1st part); What can we learn from genetic epidemiology and genomics?   

Chairs:MikeBrenner,HarvardMedicalSchoolandAnneBarton,TheUniversityofManchester

17:00–17:25Polygenicburdensoncell‐specificpathwaysunderlietheriskofrheumatoidarthritis

KazuhikoYamamoto,UniversityofTokyo

17:25–17:50Rheumatoidarthritis:theroleofenvironmentandgenes

JillNorris,Denver

17:50–18:15

Combininggeneticsandepigeneticdatatodefinekeypathogenicsinglecellpopulationsinrheumatoidarthritis

SoumyaRaychadhuri,HarvardMedicalSchool,Boston,MA

 

 

www.RAforum2017.com

18:15–18:30

Selectedabstract:TheassociationbetweenHLA‐DRB1allelesandriskofrheumatoidarthritisisinfluencedbymassivegene‐geneinteractions

Lina‐MarcelaDiaz‐Gallo,KarolinskaInstitutet,Stockholm

18:30–19:00

Discussionsonstrategicthinkingconcerningcombinationsofcohortsandbiomarkersforfuturedesignofstudiesonpathogenesis,preventionandtherapy

Discussionintroducedandledbychairpersons

19:00–21:00 ReceptionatAulaMedica

Sunday, September 24, 2017 

Session 4: Why do we get RA? (2nd part) The longitudinal course of RA 

Chairs:AnnaRudin,GothenburgUniversityandMikeHolers,UniversityofColoradoatDenver

8:30–8:55Fromtriggeringtotargeting;thelongitudinalcourseofseropositiveRA

AncaCatrina,KarolinskaInstitutet,Stockholm

8:55–9:20AutoantibodiesandtheirglycosylationduringemergenceofRA

HansUlrichScherer,LeidenUniversity

9:20–9:45ACPAs,NETsandinflammation.Newlightonneutrophils

MarianaKaplan,NIH,Bethesda,MA

9:45–10:20

PathobiologyofRheumatoidArthritis:TowardsaMolecularDefinitionandPrecisionMedicine

CosPitzalis,London

10:20–10:50 Morningbreakwithcoffeeandfruits

Session 5: Why do we get RA? (3rd part): Microbes and RA 

Chairs:ReneToes,LeidenUniversityMedicalCenterandSolbrittRantapää‐Dahlqvist,UmeåUniversity

10:50–11:15ThebacterialwaytowardsRA1:TheporphyromonasandPADpathway

KarinLundberg,KarolinskaInstitutet,Stockholm

11:15–11:40ThebacterialwaytowardsRA2:TheAggregatibacterandhypercitrullinationway.

FelipeAndrade,JohnsHopkins,Baltimore,MD

11:40–12:05RoleofthemicrobiomeinthepathogenesisofRA

XuanZhang,PekingUnionMedicalCollege

12:05–12:30DiscussionsonstrategiestounderstandthetriggersandearlystagesofRA

Discussionintroducedandledbychairpersonsfromsession4and5

12:30–13:30 Lunch

 

 

www.RAforum2017.com

Session 6: Adaptive vs. Innate immunity and mesenchymal functions as driving forces in RA 

Chairs:AndyCope,King’sCollegeLondonandSteffenGay,UniversityofZürich

13:30–13:55RoleofspecificBandTcellsimmunityinRA

VivianneMalmström,KarolinskaInstitutet,Stockholm

13:55–14:20TheBcellrepertoireinRA

DanMueller,UniversityofMinnesota

14:20–14:45

EffectsofmonoclonalantibodiesfromRApatientsoninnatefunctionsofsynovialcells

BillRobinson,StanfordUniversity,CA

14:45–15:00Selectedabstract:MonoclonalACPA‐IgGfeatureextensiveFabglycosylation

KatyLloyd,KarolinskaInstitutet,Stockholm

15:00–15:15Selectedabstract:ThemutatedRNAsplicingproteinhnrnp‐a3isanovelautoantigeninsystemicrheumaticdiseasesalinktoWarburgeffectinRA

BiankaMarklein,ChariteUniversity,Berlin

15:15–15:45HowcanwecombineunderstandingofmesenchymalandimmunefunctionsinRA?

IainMcInnes,UniversityofGlasgow

15:45–16:10DiscussionsonthedrivingforcesofRA

Discussionintroducedandledbythechairpersons

16:10–16:40 Afternoonbreakwithcoffee

Session 7: Mechanisms of pain in RA 

Chairs:CarlTuresson,LundUniversityandCodrutaFilip,PARE/EULAR

16:40–17:05PainandcognitioninRA,Whicharethecentralmechanisms?

GeorgSchett,ErlangenUniversityClinic

17:05–17:30

LongitudinaldevelopmentofpaininRA

JonLampa,KarolinskaInstitutet,Stockholm

17:30–17:55Antibody‐mediatedpain

CamillaSvensson,KarolinskaInstitutet,Stockholm

17:55–18:15 Discussionintroducedandledbythechairpersons

18:15–19:30

Posterswithpostercompetition,discussionclubsandotherinformalmeetingswithwineandcheese(posterscanbeputupduringSundayafternoonandhavetobetakendownaftertheendofthepostersession.)

Speakersdinner

 

 

www.RAforum2017.com

Monday, September 25, 2017 

 Session 8: Adaptive vs innate immunity and mesenchymal functions in RA (continued) 

Chairs:LisavanBaarsen,AMCandTBC

8:30‐8:55MFAP5playsakeyroleininvasionofFL‐likesynoviocyteinRA

HujiXu,TsinghuaUniversitySchoolofClinicalMedicine,andTheSecondMilitaryMedicalUniversity,China

8:55‐9:20Genetics,TcellspecificityandTcellregulationinRA

JaneBuckner,BenaroyaResearchInstitute,Seattle,WA

9:20–9:45HowcanadaptiveimmunefunctionsinfluencestromalcellsinRA?

CarolineOspelt,UniversityofZürich

9:45–10:00

Selectedabstract:Machinelearningusingrheumiatoidarthritissynovialgeneexpressiondatadefineshistologicfeaturesthatcorrelatewithsystemicinflammationandautoantibodies

DanaOrange,RockefellerUniversity

10:00–10:20 Discussionsintroducedandledbythechairpersons

10:20–10:45 Morningbreakwithcoffee

Session 9: Genetics and function of citrullination and PAD and other post‐translational modifications in RA 

Chairs:CarolineGrönwall,KarolinskaInstitutetandGerPruijn,RadboudUniversity

10:45‐11:00Introductiontopost‐translationalmodifications

GerPruijn,RadboudUniversity,Nijmegen

11:00–11:25TherolesofPADsinRA

HirofumiShoda,UniversityofTokyo

11:25–11:50Roleofosteoclastsinboneinflammation

HiroshiTakayanagi,UniversityofTokyo

11:50–12:05Selectedabstract:RA‐associatedantibodiestargetingposttranslationalmodificationhavedifferentosteoclastogenesispotential

AkilanKrishnamurthy,KarolinskaInstitutet,Stockholm

12:05–12:30 Discussionsintroducedandledbythechairpersons

12:30–13:45 Lunch

Session 10: Can we prevent RA? 

Chairs:Chairs:DimitrisBoumpas,BiomedicalResearchFoundationAcademyOfAthensandLarsAlfredsson,KI

13:45–14:10Trialsforprevention:Howtodoit?

TomHuizinga,LeidenUniversityMedicalCenter

14:10–14:35ModifiableriskfactorsforRA

SaedisSaevarsdottir,KarolinskaInstitutet

14:35–15:00Howtounderstandandmeasure“subjectivesymptoms”thatmatterforthepatient?

KarimRaza,UniversityofBirmingham

15:00–15:30 Afternoonbreakwithcoffee

 

 

www.RAforum2017.com

Session 11: New ways of understanding the individual’s most urgent problems and how they change over 

time; What role for e‐health 

Chairs:GerdBurmester,Charité,BerlinandIngerGjertsson,UniversityofGothenburg

15:30–15:55EmergingopportunitiesfordigitalhealthresearchinRA

WillDixon,UniversityofManchester

15:55–16:20Meetingthepatientsearlywithanapp:Whatoptionsandwhatobstacles?

SofiaErnestam,KarolinskaInstitutet

16:20–17:05Whatwillhappenwith“patientreportedoutcomes”inthefuture?

ToreKvien,OsloUniversity

17:05–17:30 Discussionintroducedandledbythechairpersonsfromsession10and11

18:30 LeaveforCityHall(busseswillleavefromoutsideAulaMedica)

19:00ThereceptionintheCityHallishostedbytheStockholmCountyCouncilandtheCityofStockholm

Tuesday, September 26, 2017 

Session 12: Future trials for prevention and curative treatments; Which strategies? 

8:30–8:55

Chairs:MarcFeldmann,UniversityofOxfordandRonaldvanVollenhoven,KIandUniversityofAmsterdam

Tolerancetherapies–willtheybecomereal?

RanjenyThomas,UniversityofQueensland

8:55–9:20TolerizingTcells

RikardHolmdahl,KarolinskaInstitutet

9:20–9:45Antigen‐specificimmunotherapy:toleranceorimmunemodulation?

DavidWraith,UniversityofBirmingham

9:45‐10:15 Morningbreakwithcoffee

10:15–10:40Newsmarttrialdesigns

JohnIsaacs,NewcastleUniversity

10:40–11:00BioelectronicMedicine:TechnologyTargetingMolecularMechanismsforTherapy

PederOlofsson,KarolinskaInstitutet

 

 

www.RAforum2017.com

Session 13: Future strategies for prevention and treatments of RA – a collaboration between academia, 

industry and health care 

chairs:LarsKlareskog(KI),MarcFeldmann(UniversityofOxford)andRonaldvanVollenhoven(KIandUniversityofAmsterdam)

11:00–12:30

Discussionwithrepresentativesfromacademia,industryandhealthcareledbythechairpersons

Perspectivesfromindustry,academiaandhealthcare;Howshouldwecombineoureffortstomakethenewgenerationofcurativeandpreventivetherapiesreal?

Confirmedguests:

AnishSuri,Janssen

AaronWinkler,Pfizer

StevenNadler,BMS

12:30–12:55AlternationsintonsilmicrobiotaassociatedwithRA

ZhanguoLi,BeijingUniversity

13:00 LunchandDepartures

Practical Information 

Posters:CanbeputupfromSundaylunchtimeandshallbetakendownSundayeveningafterthepostersession.

OurvenueisAulaMedica,Nobelsväg6,Solna(KarolinskaInstitutetcampus)

TogetherefromArlandaairport:Useacreditedtaxicompany,wewouldrecommendTaxiStockholm+468150000orTaxiKurir+468300000.ThereisalsoatrainservicetothecentralofStockholmfromArlanda;Arlandaexpress.

PublictransportisrunbySL,theyofferavarietyofticketswhichallmustbeboughtbeforeenteringthetrainorbus.Planyourtravelhere:https://sl.se/en

  

 

www.RAforum2017.com    

Last name  First name  E‐mail  Company /Organisation 

Abdelmoaty  Sally  [email protected]  Karolinska Institutet 

Alemo Munters  Li  [email protected] Swedish Reumatism Association

Alfredsson  Lars  [email protected] Karolinska Institutet 

Almgren  Malin  [email protected]  Karolinska Institutet 

Alunno  Alessia  [email protected]  University of Perugia 

Andersson  Maria  [email protected]  AbbVie AB 

Andersson  Åsa  [email protected]  Halmstad University  

Andrade  Felipe  [email protected] Johns Hopkins UniversityAranda‐Guillén  Maribel  [email protected]  Karolinska Institutet 

Arkema  Elizabeth  [email protected]  Karolinska Institutet 

Askling  Johan  [email protected]  Karolinska Institutet 

Azali  Payam  [email protected]  Danderyds sjukhus Stockholm 

Baharpoor  Azar  [email protected]  Karolinska Institutet 

Barton  Anne  [email protected] University of Manchester

Berg  Louise  [email protected]  Karolinska Institutet 

Bergman  Thomas  [email protected]  Karolinska Institutet 

Bernmalm  Anna  [email protected]  AbbVie 

Bersellini Farinotti  Alex  [email protected] 

Karolinska Institutet 

Bi  Li Qi  [email protected] China‐Japan Union Hospital of Jinlin 

Bing  Liu  [email protected]  Henan People's Hospital 

Boddul  Sanjay  [email protected]  Karolinska Institute 

Bokarewa  Maria  [email protected]  University of Gothenburg 

Brenner  Michael  [email protected] Medical School/ Brigham & Women's Hospital

Brink  Mikael  [email protected]  Umeå University 

Bucher  Sara sara.magnusson‐[email protected]  Universitetssjukhuset Örebro 

Buckley  Christopher  [email protected] Universities Oxford and Birmingham 

Buckner  Jane  [email protected] Benaroya Research Institute

Bäckdahl  Liselotte  [email protected]  Karolinska Institutet MBB 

  

 

www.RAforum2017.com 

Bäcklund  Johan  [email protected]  Karolinska Institute 

Cai  Weiwei  [email protected]  Karolinska Institute 

Cai  Xiaoyan  [email protected] Guangzhou First Municipal Hospital 

Carlberg  Konstantin  [email protected]  Scilifelab 

Carlberg  Renée  [email protected]  Bristol‐Myers Squibb 

Carlens  Cecilia  [email protected]  Karolinska Universitetssjukhuset 

Cerqueira  Cátia    Karolinska Institutet Chatzidionysiou  Katerina  [email protected]  Karolinska Institutet 

Chemin  Karine  [email protected]  Karolinska Institutet 

Chen  Jinwei  [email protected]  second xiangya hospital 

Chen  Pei  [email protected]  CTS MICE SERVICE CO LTD 

Chen  Szu‐Ying  szu‐[email protected]  Karolinska Institutet 

Chen  Weiqian  [email protected]  the First Affiliated Hospital, Coll 

Chen  Zhu  [email protected]  ANHUI PROVINCIAL HOSPITAL 

Cope  Andrew  [email protected]  King's College London 

Cui  Yang  [email protected]  Guangdong General Hospital 

Curman  Charlotte  [email protected]  Pfizer 

De Pablo  Paola  [email protected]  University of Birmingham 

De Vries  Niek  [email protected]  AMC/University of Amsterdam 

Degerlund  Helene  [email protected]  Roche AB 

Deleskog  Anna  [email protected]  Sandoz A/S 

Dellmark  Yvonne  [email protected]  Karolinska Universitetssjukhuset 

Deng  Guo‐Min  [email protected]  Nanjing Medical University 

Diaz  Lina  [email protected]  Karolinska Institutet 

Dimitris  Boumpas  [email protected]   BRFAA 

Ding  Jian  [email protected]  Lihuili hospital of Ningbo City 

Dixon  William  [email protected]  The University of Manchester 

Dong  Xiaoying  [email protected]  TJMU GENERAL HOSPITAL 

  

 

www.RAforum2017.com 

Dorph  Christina  [email protected]  Karolinska university hospital 

Dubnovitsky  Anatoly  [email protected]  Karolinska Institutet 

Edvinsson  Helene  [email protected]  Pfizer 

Egnell  Maud  [email protected]  Eli Lilly 

Ekström  Tomas  [email protected]  Karolinska Institutet 

Elfverson  Hanna  [email protected]  Thermofisher Scientific 

Emamikia  Sharzad  [email protected]  CLINTRID 

Engström‐Laurent  Anna  anna.engstrom‐[email protected] 

Dept.Public Health and Clinical Med 

Enman  Yvonne  [email protected]  Reumatikerförbundet 

Erlandsson Harris  Helena  [email protected]  Karolinska Institutet 

Ernestam  Sofia  [email protected]  SLSO / Karolinska Institutet 

Fan  Xiaoyun  [email protected] The Affiliated No.1 Hospital of BMC 

Fasth  Andreas  [email protected]  Novartis 

Fei  Guozhong  [email protected]  Karolinska Institutet 

Felix  Nathan  [email protected]  Janssen R&D 

Fernandez  Gonzalo  [email protected]  Karolinska Institutet 

Fischer  Anita  [email protected]  Medical University of Vienna 

Fogdell‐Hahn  Anna  anna.fogdell‐[email protected]  Karlinska Institutet 

Foxryd  Preben  [email protected]  Pfizer 

Fu  Xiao  [email protected]  Univerzita Karlova 

Fu  Xinli  [email protected] AffiliatedHospitaShandongUniversity 

Gammeltoft Gerwien  Jens  [email protected]  Eli Lilly 

Gao  Guanmin  [email protected]  the First Affiliated Hospital of Zh 

Gay  Steffen  [email protected] Dept Rheumatology Univ Hospital Zurich, Switzerland 

Ge  Changrong  [email protected]  Karolinska Institutet 

Gerstner  Christina  [email protected]  Karolinska Institutet 

Gjertsson  Inger  [email protected] Göteborgs Universitet, Reumatologi 

Goodyear  Carl  [email protected]  University of Glasgow 

Grönwall  Caroline  [email protected]  Karolinska Institutet 

Gu  Jieruo  [email protected]  Third Affiliated Hospital of Sun Ya 

  

 

www.RAforum2017.com 

Guenter  Steiner  [email protected]  Medical University Vienna 

Gunnarsson  Karin  [email protected]  Karolinska Univerity Hospital 

Guo  Jianping  [email protected]  Peking University 

Gülfe  Anders  [email protected]  Lund University, Rheumatology 

Hafström  Ingiäld  [email protected]  Karolinska Institutet 

Hagert  Cecilia  [email protected]  University of Turku 

Hallström  Magnus  [email protected] Sahlgrenska Universitetssjukhuset 

Hambardzumyan  Karen  [email protected]  Karolinska Institutet 

Hansson  Monika  [email protected]  Karolinska Institutet 

Harju  Anders  [email protected] Handelsbolag, Harpo HB 969636‐6286  

He  Lan  [email protected]  UMICE 

He  Yibo  [email protected]  Karolinska Institutet 

Hedenstierna  Louise  [email protected]  Karolinska Institutet 

Heiberg  Turid  [email protected]  Oslo University Hospital 

Hellgren  Karin  [email protected] Rheumatology Unit  Karolinska University Hospital 

Hermanrud  Christina  [email protected]  Karolinska Institutet 

Hernandez  Susana  [email protected]  Karolinska Institutet 

Hetland  Merete  [email protected]  Rigshospitalet ‐ Glostrup 

Hoffmann  Markus  markus.hoffmann@uk‐erlangen.de University of Erlangen‐Nuremberg 

Holers  Michael  [email protected] University of Colorado Division of Rheumatology 

Hou  Yong  [email protected] Peking UnionMedicalCollege Hospital 

Houtman  Miranda  [email protected]  Karolinska Institutet 

Hu  Dawei  [email protected] Renji Hospital Affiliated to Shangh 

Hu  Xiaobin  [email protected]  Hospital of traditional  Medine 

Huang  Anbin  [email protected]  Wuhan Union Hospital 

Huang  Yanhong  [email protected]  Beijing  Jishuitan Hospital 

  

 

www.RAforum2017.com 

Huizinga  Tom  [email protected]  Leiden University Medical Center Hultgård Ekwall  Anna‐Karin  [email protected]  Sahlgrenska Akademin 

Idborg  Helena  [email protected]  Karolinska Institutet 

Ilar  Anna  [email protected]  Karolinska Institutet 

Illescas Rodriguez  Vera  [email protected] 

Isaacs  John  [email protected]  Newcastle University 

Israelsson  Lena  [email protected]  Karolinska Institutet 

Jakobsson  Per‐Johan  per‐[email protected]  Karolinska Institutet 

James  Jaime Rose  [email protected]  Karolinska Institute 

Jenning  Madeleine  [email protected]  Charité Universitätsmedizin Berlin

Ji  Lanlan  [email protected]  Peking University First Hospital 

Jia  Ru Lin  [email protected] Peking University People's Hospital 

Jiajia  Liu  [email protected] Peking University People's Hospital 

Jiang  Long  [email protected]  Karolinska Institutet 

Jimenez‐Andrade  G Yanek  [email protected]  Karolinska Institutet 

Jiping  Wu  [email protected]  Dongguan People's Hospital 

Johannesson  Martina  [email protected]   Karolinska Institutet 

Johansson  Eva C.  [email protected] PRP / Swedish Rheumatism Association 

Johansson  Linda  [email protected]  Umeå Universitet 

Johnston  Lynsey  [email protected]  University of Leeds 

Jun  Wu  [email protected]  Huitian Pharmaceutical Co.,Ltd 

Jurczak  Alexandra  [email protected]  Karolinska Institutet 

Kampstra  Sanne  [email protected]  Leiden University Medical Center 

Kaplan  Mariana  [email protected]  National Institutes of Health 

Karlsson Marie‐Louise  marie‐[email protected]  Karolinska Universitetssjukhuset 

Kastbom  Alf  [email protected]  Linköping University Hospital 

Kharlamova  Anastasia  [email protected]  Karolinska Institutet 

  

 

www.RAforum2017.com 

Kisten  Yogan  [email protected]  Karolinska Institutet 

Klareskog   Lars  [email protected]  Karolinska Institutet 

Klein  Kerstin  [email protected]  University Hospital Zurich 

Klosinska Linder  Maria  [email protected] 

Reumatologklin i Sunderby Sjukhus 

Knutsson  Susanna  [email protected]  Bristol‐Myers Squibb 

Koen  Vos  [email protected] Academic Medical Center, Amsterdam 

Kokkonen  Heidi  [email protected]  Umeå University 

Korotkova  Marina  [email protected]  Karolinska Institutet 

Kozhukh  Genadiy  [email protected]  KI 

Krishnamurthy  Akilan  [email protected]  Karolinska Institutet 

Krock  Emerson  [email protected]  Karolinska Institutet 

Krönke  Gerhard  gerhard.kroenke@uk‐erlangen.de  University of Erlangen 

Kvien Tore Kristian  [email protected] 

Dept of Rheumatology,  Diakonhjemmet Hospital 

Kämpe  Olle  [email protected]  Karolinska Institutet 

L Schultzberg  Carin  [email protected]  AbbVie AB 

Lampa  Jon  [email protected]  Dept of Rheumatology, KI 

Larsson  Esbjörn  [email protected]  Eli Lilly 

Le Bars  Manuela  [email protected]  Janssen  

Le Maitre  Erwan  [email protected]  Karolinska Institutet 

Lend  Kristina  [email protected]  Karolinska Institutet 

Leng  Nan  [email protected]  XIJING HOSPITAL 

Li  Caifeng  [email protected]  Beijing Children's Hospital 

Li  Cunyan  [email protected]  The People’s Hospital of Hunan  

Li  Fen  [email protected] THE SECOND XIANGYA HOSPITAL  OF CSU 

Li  Hong  [email protected]  Linyi People’s Hospital  

Li  Qin  [email protected]  Qinghai Hospital of traditional Chi 

Li  Shiguang  [email protected]  PFIZER INVESTMENT CO., LTD 

Li  Taotao  [email protected]  Karolinska Institutet 

Li  Xia  [email protected]  Dalian Medical University 

Li  Xiaomei  [email protected]  Anhui provincial hospital 

  

 

www.RAforum2017.com 

Li  Yinong  [email protected]  Affiliated Fujian Wujing Hospital 

Li  Yuhui  [email protected] People's Hospital of Peking Univers 

Li  Zhanguo  [email protected]  UMICE 

Liao  Hua  [email protected]   Beijing Anzhen Hospital  

Lindberg  Tülay  [email protected]  Karolinska Institutet 

Lindencrona  Jan Alvar  [email protected]  Karolinska Institutet 

Lindgren  Inger  [email protected]  MedicILAB 

Lindqvist  Elisabet  [email protected]  Skånes Universitetssjukvård 

Lindqvist  Joakim  [email protected]  Karolinska Institutet 

Lindroos  Eva  [email protected]  Karolinska Institutet 

Lindskog  Hans  [email protected]  Bristol‐Myers Squibb 

Ling  Guanghui  [email protected]  The Second Xiangya Hospital 

Liu  Yanying  [email protected] Peking University People's Hospital 

Lloyd  Katy  [email protected]  Karolinska Institutet 

Lu  Jing  [email protected] The  First Hospital of China Medica 

Lu  Yue Wu  [email protected]  BEIJING CHAOYANG HOSPITAL 

Lubberts  Erik  [email protected] Erasmus MC, University Medical Cent 

Lundberg  Ingrid  [email protected]  Karolinska Institutet 

Lundberg  Karin  [email protected]  Karolinska Institutet 

Lundblad  Per  [email protected] Reumabulletinen Mediahuset i Gbg AB 

Lundell  Anna‐Carin  anna‐[email protected]  Gothenburg University 

Luo  Jing  [email protected]  Shanxi Medical University 

Lv  Xing  [email protected]  China 

Lydén  Malin  [email protected]  Landstinget Kalmar län,  

Lyu  Liangjing  [email protected]  Renji Hospital 

Lönnblom  Erik  [email protected]  Karolinska Institute 

Maini  Ravinder  [email protected] Kennedy Institute of Rheumatology 

Maldonado  Michael  [email protected]  Bristol‐Myers Squibb 

Mark  Linda  [email protected]  Sahlgrenska Academy 

Marklein  Bianka  [email protected] Charite/ Rheumatolog. Forschungslab 

Martinsson  Klara  [email protected]  Linköping University 

  

 

www.RAforum2017.com 

Mathsson Alm  Linda  linda.mathsson‐[email protected]  Thermo Fisher Scientific 

Mcinnes  Iain  [email protected]  University of Glasgow 

Melasniemi  Jonna  [email protected]  Bristol‐Myers Squibb 

Mia Muhammad Sohel  [email protected]  Karolinska Institutet 

Moberg  Martin  [email protected]  Eli Lilly 

Morado Urbina 

Carlos Eduardo  [email protected]  

Karolinska Institutet 

Mueller  Daniel  [email protected] University of Minnesota Medical School 

Möller  Petronella  [email protected]  Sandoz A/S 

Nadler, Phd  Dr. Steven  [email protected]  Bristol Myers Squibb 

Nascimento  Diana  [email protected] Karolinska Institutet 

Neofytou  Christina  [email protected]  Karolinska Institutet 

Neregård  Petra  [email protected]  Centrum för reumatologi 

Niklason  Anders  [email protected]  Pfizer 

Nilesh  Agalave  [email protected]  Karolinska Institutet 

Nisell  Ralph  [email protected]  Centrum för Reumatologi 

Nordström  Inger  [email protected]  Gothenburg University 

Norin  Ulrika  [email protected]  Karolinska Institutet 

Norkko  Julia  [email protected]  Karolinska Institutet 

Norris  Jill  [email protected]  Colorado School of Public Health 

Nortamo  Pekka  [email protected]  Eli Lilly 

Notarnicola  Antonella  [email protected]  Karolinska Institutet 

Nyman  Ulf  [email protected]  Utsikten 

Oke  Vilija  [email protected]  Karolinska University Hospital 

Olofsson  Peder  [email protected]  Karolinska Institutet 

Opava  Christina  [email protected]  Karolinska institutet 

Orange  Dana  [email protected]  Rockefeller University 

Ospelt  Caroline  [email protected]  University of Zurich 

Ossipova  Elena    Karolinska Institutet 

Ota  Mineto  mioota‐[email protected]  The University of Tokyo 

Padyukov  Leonid  [email protected]  Karolinska Institutet Palm Gustafsson  Ulrika  ulrika.palm‐[email protected]  Bristol‐Myers Squibb 

Pan  Yunfeng  [email protected] THE THIRD AFFILIATED HOSPITAL,SYSU 

Panda Sudeepta Kumar  [email protected]  Karolinska Institute 

  

 

www.RAforum2017.com 

Parodis  Ioannis  [email protected] 

Peng  Linyi  [email protected] Peking Union Medical College Hospit 

Pihl  Maria  [email protected]  Eli Lilly 

Pitzalis  Costantino  [email protected] 

William Harvey Research Institute Queen Mary University of London 

Pratt  Arthur  [email protected]  Newcastle University 

Pruijn  Ger  [email protected]  Radboud University 

Qiang  Guo  [email protected]  Shanghai Renji Hospital 

Ramsköld  Daniel  [email protected]  Karolinska Institutet 

Rantapää Dahlqvist  Solbritt  [email protected]  Umeå University 

Rao  Navin  [email protected]  Janssen Research & Development 

Raychaudhuri  Soumya  [email protected]  Brigham and Women's Hospital 

Raza  Karim  [email protected]  University of Birmingham 

Reimertz  Claus  [email protected]  Sanofi 

Réthi  Bence  [email protected]  Karolinska Institutet 

Ringh  Mikael  [email protected]  Karolinska Institutet 

Robinson  William  [email protected]  Stanford University 

Rossides  Marios  [email protected]  Karolinska Institutet 

Rostvall  Gloria  [email protected]  Karolinska Institutet 

Rudin  Anna  [email protected] The Sahlgrenska Academy at  University of Gothenburg 

Rudjito  Resti  [email protected]  Karolinska Institutet 

Rydell  Emil  [email protected] Institutionen för Kliniska Vetenska 

Saevarsdottir  Saedis  [email protected]  Karolinska Institutet 

Sahlström  Peter  [email protected]  CMM/Charité 

Salter  Hugh  [email protected]  Anocca AB 

Sandman  Maria  [email protected]  Eli Lilly 

Sardar  Samra  [email protected]  University of Copenhagen 

Saxena  Amit  [email protected]  Karolinksa Institutet 

Scheel‐Toellner  Dagmar  [email protected]  University of Birmingham 

Scherer  Hans Ulrich  [email protected]  Leiden University Medical Center 

  

 

www.RAforum2017.com 

Schett  Georg  georg.schett@uk‐erlangen.de  Universitätsklinikum Erlangen 

Seddighzadeh  Maria  [email protected]  MSD 

Seidman  Jade  [email protected]  Synergy Medical  

Sener  Zeynep  [email protected]  Karolinska Institute 

Shang  Mingmei  [email protected]  KI 

Shen  Hui  [email protected] The First Hospital Of China Medical 

Sherina  Natalia  [email protected]  Karolinska Institutet 

Shi  Yunzhen  [email protected]  Guang Dong General Hospital 

Shoda  Hirofumi  shoda‐[email protected]  Tokyo University 

Sippl  Natalie  [email protected]  Karolinska Institutet 

Sizhao  Li  [email protected]  China‐Japan Friendship hospital 

Sjöholm  Louise  [email protected]  KI 

Skougaard Nielsen  Marie  [email protected]  The Parker Institute 

Skriner  Karl  [email protected]  Charite 

Steen  Johanna  [email protected]  Karolinska Institutet 

Stenberg  Jan  [email protected]  Biogen 

Stevenaert  Frederik  [email protected]  Janssen Pharmaceutical cies 

Stolt  Patrik  [email protected]  To be announced 

Studenic  Paul  [email protected]  Medical University of Vienna 

Stålesen  Ragnhild  [email protected]  Karolinska Institutet 

Su  Yin  [email protected] Peking University People's Hospital 

Sun  Erwei  [email protected]  Southern Medical University 

Sun  Meng  [email protected]  Karolinska Institutet 

Sun  Mingshu  [email protected]  The Affiliated Hospital of Qingdao 

Sun  Xiaolin  [email protected] Peking University People's Hospital 

Sundberg  Erik  [email protected]  Karolinska 

Suri  Anish  [email protected]  Janssen, J&J 

Svensson  Camilla  [email protected]  Karolinska Institutet 

Svenungsson  Elisabet  [email protected]  Karolinska Institutet 

Söderbergh  Annika  [email protected]  Örebro Universitetssjukhus 

Takayanagi  Hiroshi  [email protected]  The University of Tokyo 

  

 

www.RAforum2017.com 

Tang  Yanchun  [email protected]  YTYHD HOSPITAL 

Tao  Jinhui  [email protected]  Anhui Provincial Hospital 

Tengelin  Sara  [email protected]  Sandoz A/S 

Thomaidi  Eleni  [email protected]  Universitetssjukhuset Örebro 

Thomas  Ranjeny  [email protected]  University of Queensland 

Tidblad  Liselotte  [email protected]  Karolinska Universitetssjukhuset 

Tideström  Margareta  [email protected]  Karolinska institutet 

Tinna Ängeby‐Möller  [email protected]  

Karolinska Institutet 

Tjärnlund  Anna  [email protected]  Karolinska Institutet / UCB 

Toes  René  [email protected]  Leiden University Medical Center 

Trollmo  Tina  [email protected]  Roche AB 

Tsuchiya  Haruka  [email protected]  The University of Tokyo 

Tu  Shenghao  [email protected]  Tongji Hospital 

Turcinov  Sara  [email protected]  Karolinska Institutet 

Turesson  Carl  [email protected]  Skåne University Hospital 

Urbonaviciute  Vilma  [email protected]  Karolinska Institutet 

Vaartjes  Danielle  [email protected]  Karolinska Institutet 

Waldenlind  Kristin  [email protected]  Karolinska Institutet 

Wallman  Thomas  [email protected]  Sanofi Genzyme 

Van Baarsen  Lisa  [email protected]  Academic Medical Center 

Van Schie  Karin  [email protected]  LUMC 

van Vollenhofen  Ronald  [email protected] 

Karolinska Institutet/ University of Amsterdam 

Wang  Bing  [email protected]  Dalian Medical University 

Wang  Jibo  [email protected]  The Affiliated Hospital of QDU 

Wang  Yan  [email protected]  Karolinska Institutet 

Wang  Yuxuan  [email protected] Peking University People's Hospital 

Wenfeng  Tan  [email protected]  Jiangsu Province Hospital 

Wermeling  Fredrik  [email protected]  Karolinska Institutet 

  

 

www.RAforum2017.com 

Westerlind  Helga  [email protected]  Karolinska Institutet 

Wick  Cecilia  [email protected]  Karolinska Institutet 

Wigerblad  Gustaf  [email protected]  Karolinska Institutet 

Winkler  Aaron  [email protected] Pfizer Inflammation and Immunology 

Vinko  Palada  [email protected]  Karolinska Institutet 

Volkov  Mikhail  [email protected]  Leiden University Medical Center 

Wollheim  Frank  [email protected]  Lund University 

Wraith  David  [email protected]  University of Birmingham 

Wu  Huaxiang  [email protected]  The 2nd Affiliated Hospital 

Wu  Jianxiong  [email protected]  Guangzhou Orthopedic Hospital 

Wu  Lijun  [email protected] PEOPLE'S HOSPITAL OF XINJIANG UAR 

Wållberg Jonsson  Solveig  [email protected]  Umeå Universitet 

Wänkkö  Katarina  [email protected] Reumatomogen Sunderby sjukhus 

Xu  Bingze  [email protected]  Karolinska Institute 

Xu  Guoli  [email protected]  Peking University People's Hos 

Xu  Haijun  [email protected] AbbVie Pharmaceutical Trading(SH) 

Xu  Jianhua  [email protected] The Affiliated No.1 Hospital of AMU 

Xu  Liang  [email protected] Yijishan hospital of Wannan medical 

Yamamoto  Kazuhiko  [email protected] RIKEN Center for Integrative  Medical Sciences 

Yan  Bing  [email protected] WestChinaHospitalSichuanUniversity 

Yang  Min  [email protected]  West China hospital 

Yang  Min  [email protected]  Karolinska Institutet 

Yao  Zhongqiang  [email protected]  Peking University Third Hospital 

Yeremenko  Nataliya  [email protected]  Academic Medical Center 

Yongkang  Wu  [email protected] WestChinaHospital SichuanUniversity 

  

 

www.RAforum2017.com 

 

Zamout  Petros  [email protected]  Reumatologkliniken, USÖ 

Zeng  Pingling  [email protected]  Karolinska Institute 

Zhan  Feng  [email protected]  Hainan General Hospital 

Zhang  Jing  [email protected] Sichuan Provincial People’s Hospita 

Zhang  Juan  [email protected] Lanzhou University Second Hospital  

Zhang  Ning  [email protected] Shengjing Hospital Of China Medical 

Zhang  Xuan  [email protected] PEKING UNION MEDICAL  COLLEGE HOSPITAL 

Zhang  Xuewu  [email protected] PEKING UNIVERSITY  PEOPLE’S HOSPITAL 

Zhao  Jinkang  [email protected]  XI‐Jing Hospital 

Zheng  Baolin  [email protected]  Foshan Hospital of traditional 

Zheng  Zhaohui  [email protected] The Affiliated No.1 Hospital of ZZU 

Zhong  Jianghong  [email protected]  Karolinska Institutet 

Zhou  Diana  [email protected] Karolinska Sjukhuset,  dep. of Medicine 

Zhou  Hang  [email protected]  Beijing Friendship Hospital 

Zhou  Jiaxin  [email protected] Peking Union MedicalCollegeHospital 

Zhu  Xiaochun  [email protected]  The First Affiliated Hospital  

Zhuoli  Zhang  [email protected]  Peking University First Hospital 

Östberg  Therese  [email protected]  Sanofi Genzyme 

1. NEW PROTEIN ARRAY TECHNOLOGY IDENTIFIES RITUXIMAB TREATED NON RESPONDER RHEUMATOID ARTHRITIS PATIENTS ARE GENERATING A NEW AUTOANTIBODY REPERTOIRE

Z. Konthur1, M. Wiemkes 2, T. Häupl2, G. Burmester1, K. Skriner2,*

1Max Planck Institute , Max Planck Institute , 2Department of Rheumatology and Clinical Immunology, CHARITÉ UNIVERSITY MEDICINE, Berlin, Germany

Objectives: Rituximab (RTX) has shown clinical efficacy but up to 40 % of RTX treated rheumatoid arthritis (RA) patients are poor responders (Ann-Rheum-Dis. 2005 Feb;64(2):246-52) and the commonly used RA biomarkers (RF/ACPA) are poor predictors for therapy response. In this study the autoantibody repertoire analysed on protein macorarrays from RA patients under RTX treatment was correlated to clinical DAS28 response.

Methods: Screening of RA sera was conducted on 37.830 unique human proteins on protein marcoarrays (http://www.engine-gmbh.de) with sera taken before and 24 weeks after treatment. The autoantibody response of different immunoglobulin classes IgD, IgA, and IgG was recorded and bioinformatically evaluated. Response was determined according to DAS28 criteria. DAS 28 scores in the responder group before treatment was from 5.4 – 7.8 and in the non-responder group 5,6 – 6,8. We analyzed 26 RA patient sera (9 responder, 7 non-responder and 10 patients with blinded response classification) investigated the data of found autoantigens in-silico and by hierarchical clustering

Results: In the cohort of 26 patients 1292 different autoantigens (IgD,IgA,IgG) were detected. Using protein array we investigated clusters of autoantigen responses that disappeared or developed during RTX treatment of RA patients. RA autoantigenic patterns before and 6 month after RTX treatment were patient-specific and no relevant autoantigenic cluster was found that was shared between patients or associated with response. However, RTX reduced the repertoire of autoantibodies after 24 weeks of treatment in the tested RA patient cohort on average by 60%. RA patients which do not respond are generating on average 63% new autoantibodies. In good responders to RTX only 5,5% (+/-3%) new autoantibodies can be detected. The IgA and IgG autoantibody repertoire in the serum after 24 weeks of RTX treatment is reduced (IgA: 41%, IgG :31%) in good responders whereas it is increased (IgA: 1,3%, IgG: 24%) in non responders to RTX.

Conclusions: After 6 month of RTX treatment the autoantibody repertoire in all good responding RA patients is reduced and non responders to RTX change their autoantibody repertoire directed against new but patient specific antigens. The fast rebuilding of functional B cells is only detected in non-responders to rituximab

RA-associated antibodies targeting post translational modification have different osteoclastogenetic potential

Akilan Krishnamurthy1, Johanna Steen1, Caroline Grönwall1, Philip Titcombe1, Gustaf Wigerblad2, Camilla Svennson2, Heidi Wähämaa1, Vivianne Malmström1, Bence Rethi1 and Anca I Catrina1 1Rheumatology unit, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden, 2 Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden Background:

Some but not all antibodies against citrullinated modified proteins (ACPA) promote osteoclastogenesis and bone destruction in vitro and in vivo. We aimed to investigate the ACPA specificity pattern that is related to this effect and if this effect is limited to ACPA or encompasses also other RA-associated antibodies.

Methods:

Polyclonal ACPA IgG and IgGs others than ACPA were obtained from the peripheral blood of RA patients by purification on a G column followed by an anti-CCP2 column. Monoclonal ACPA, anti-

MDA and rheumatoid factor (RF) IgGs were generated from either single plasma cells isolated from the synovial fluid or tetramer-positive sorted single B-cell isolated from the plasma of RA patients. Osteoclasts were generated from CD14+ monocytes of healthy individuals or bone marrow cells of Fc gamma III or Fc gamma chain knockout mice, in the presence or absence of polyclonal ACPA, monoclonal antibodies (ACPA, and anti-MDA antibodies and RF) and IgG controls. TRAP positive multinucleated cells were counted and bone erosion assay was done in parallel.

Results:

Polyclonal ACPA increased osteoclastogenesis, by a fold of 1.6±0.03One out of 4 tested plasma cell derived monoclonals ACPAs and one out of the five tested tetramer positive B-cell derived monoclonals have similar effects (with a fold increase of 1.63 ±0.15 for the plasma cell derived antibody and 1.4± 0.16 for the tetramer positive B-cell derived) have similar OC effects. Two additional tetramer positive B-cell derived monoclonals inhibited osteocalstogenesis while the remaining had no significant effect. All monoclonal ACPA were relatively highly cross-reactive to several citrullinated epitopes but not to native arginine peptides. Anti-MDA monoclonals antibodies displaying somatic hypermutations and low reactivity had significant in vitro functional properties and enhanced osteoclastogenesis (fold increase of for one antibody 4.0±0.76 and fold increase of for the second one 2.3±0.2), while the natural antibody related high-reactivity anti-MDA antibody did not. Anti MDA antibodies had no cross reactivity to other antigen modifications such as citrullination or carbamylation. Monoclonal RF had no direct effect on ostecoalstogenesis but were able to significantly increase ACPA-mediated osteoclastogenesis (fold increase of 1.68 ±0.03 for ACPA alone and for the combination of ACPA and RF 3.15±0.24). Dimeric Fab fragments of polyclonal ACPA increased OC numbers by a fold of 1.78, suggesting that epitope recognition is involved in the observed osteocalstogenetic effect of ACPA. Interestingly however while ACPA increased osteoclastogenesis from bone marrow precursors of wild type mice, it had no effect on the bone marrow precursors of the Fc gamma chain knockout and Fc gamma III mice bone marrow samples, implying a more complex mechanism than epitope recognition alone that involves Fc receptors.

Conclusion:

We demonstrate that RA-associated antibodies targeting different post translational modifications have the capacity to increase osteoclastogenesis while others have not. The mechanism is mediated through both Fc dependent and independent mechanisms.

2. BOB.1 INDUCES COSTIMULATORY CAPACITY OF B CELLS DURING T CELL-

DEPENDENT ACTIVATION

Levels M.J1,2,3, Fehres C.M1,2,3, Germar K1,2,3, van Uden N. O. P1,2,3, Blijdorp I.C.J1,2,3, Bakker A.Q4,

O'Toole T.G5, Spits H,4,6, Baeten D1,2,3, Yeremenko N.G1,2,3

1Academic Medical Center/University of Amsterdam, 2Agendia BV, Clinical Immunology and

Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands,

2Amsterdam Rheumatology and immunology Center, Amsterdam, Netherlands, 3Experimental

Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, Netherlands, 4AIMM

Therapeutics, Amsterdam, Netherlands, 5 Department of Molecular Cell Biology and Immunology,

Vrije Universiteit Medical Center, Amsterdam, The Netherlands, 6Department of Cell Biology and

Histology, Academic Medical Centre/University of Amsterdam, Amsterdam, Netherlands

Background: B cells play an important pathogenic role in RA. Although it remains unclear what

molecular mechanisms guide or control pathogenic B cells during joint inflammation, the production

of autoantibodies and the presence of ectopic lymphoid structures in the RA synovium suggest that

local players in the synovial tissue may contribute to autoimmune tissue inflammation. Recently we

identified the transcriptional co-activator BOB.1 as specifically overexpressed in RA synovium, where

its levels strongly correlated with the presence of germinal centers (GCs). In accordance with human

data, mice lacking functional BOB.1 failed to mount GC response and were resistant to the

experimental model of RA. In this study we investigated whether increased levels of BOB.1 impact the

phenotype and function of B cells in a GC-like environment.

Methods: cDNA encoding BOB.1 was introduced into B cells isolated from peripheral blood or tonsils

by retrovirus-mediated gene transfer. The phenotype, activation, survival, proliferation and BCR

signaling were analyzed by FACS.

Results: Human B cells cultured with CD40L and IL-21 rapidly differentiate to antibody-producing

plasma cells, a process that is accompanied by decreased expression of CD20 and CD22 and an

increased expression of CD38 and CD27. In contrast, cells transduced with BOB.1 retained CD20 and

CD22 expression and were CD27intermediateCD38intermediate. Accordingly, the percentage of plasma cells

was significantly lower in BOB.1-overexpressing cells confirming that high levels of BOB.1 suppress

plasma cell differentiation. Further analysis revealed that BOB.1-transduced memory B cells showed

more rapid Ca2+ mobilization following BCR stimulation and expressed higher levels of CD40, CD80

and PD-L2 costimulatory receptors involved in B-T cell interactions and critical for the development of

T cell-mediated antibody responses. To address the role of BOB.1 on BCR-mediated antigen binding

and processing we made use of an influenza‐specific B cell clone capable of binding and internalizing

H1 antigen. Remarkably, overexpression of BOB.1 in these cells resulted not only in the elevated

expression of MHC class II, CD40 and CD86 molecules, but also in a faster binding of H1 antigen

suggesting that BOB.1 can control antigen presentation strength.

Conclusions: These data suggest that increased levels of BOB.1 in B cells during T cell-dependent

responses suppress plasma cell differentiation and impact BCR signaling strength, BCR-mediated

antigen processing and expression of costimulatory molecules. Whether it results in the broadening

and maturation of the autoimmune response and in survival of autoreactive B cells in RA joint is

currently under investigation.

3. High-throughput sequencing of auto-antigen specific antibodies in RA patients

Yan Wang, Katy Lloyd, Uta Hardt, Vivianne Malmström, Karin Lundberg, Lars Klareskog and Caroline Grönwall

Dept. of Medicine, Rheumatology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden

The presence of autoantibodies is a hallmark for many autoimmune diseases, yet exactly how autoreactive B cells are first generated is still unclear. Single-cell sequencing techniques have been previously used to analyse ACPA-secreting B cells, however higher-throughput analysis is required to

explore how autoreactivities against citrullinated antigens first originate in ACPA seropositive RA. Next-generation sequencing enables a comprehensive view of the human B cell repertoire for the first time. The high error-rate of this technology limited its application for the accurate analysis of mutations within antibody repertoires, however this has recently been resolved by attaching molecular barcodes to individual mRNA. ACPA-positive B cells isolated from RA patients can be too many in number for single-cell sequencing techniques, but too few for currently described molecular barcoding methods. To overcome this issue, we have developed a method to generate cDNA library featuring molecular barcodes from a hundred to a few thousand B cells. The heavy chain and light chain antibody gene will be amplified from this molecular-barcoded library and will be sequenced using Illumina MiSeq sequencing. The sequencing results will allow us to analyse the isotype, mutation rate and antigen-selection of autoreactive B cells in an error-free manner, owing to the use of molecular barcodes. This high-throughput method is a powerful tool to study B cell repertoires, and we hope that analysing somatic hypermutations within the antibody variable genes will allow us to track the maturation of autoreactive B cells.

4. Number and type of ACPA fine specificities are correlated to High resolution computed tomography parenchymal lungs changes in patients with early untreated rheumatoid arthritis.

Vijay Joshua1, Aase Haj Hensvold1, Gudrun Reynisdottir1, Monika Hansson1, Martin Cornillet2, Leonor Nogueira2, Guy Serre2, Sven Nyren3, Reza Karimi4, Anders Eklund4, Carl Magnus Sköld4, Johan Grunewald4, Katerina Chatzidionysiou1* and Anca I. Catrina1*

1 Rheumatology Unit, Department of Medicine, Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden

2 Unité Différenciation Épithéliale et Autoimmunité Rhumatoïde, Unité Mixte de Recherche 1056, INSERM – Université de Toulouse, Toulouse, France

3 Department of Radiology, Karolinska University Hospital, Stockholm, Sweden.

4 Division of Respiratory Medicine, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden

Background

Airways abnormalities that are consistent with inflammation are common in anti-CCP2 positive subjects without inflammatory arthritis. Anti-CCP2 antibodies are associated with HRCT parenchymal lung abnormalities in patients with early RA. This study aims to examine the association between ACPA fine specificities and HRCT lung changes in an early RA cohort and to asses these changes after 6 months.

Methods

Patients (n=106) with newly diagnosed RA according to the ACR 1987 criteria and naïve to treatment with oral glucocorticoids or DMARDs were included. HRCT was performed in order to assess the presence parenchymal (nodules, ground-glass opacities, opacities, fibrosis, emphysema) and airway abnormalities (bronchiectasis, air trapping, air wall thickening). EliA system (Phadia) was used to detect RF IgA and IgM, anti-CCP2 IgA and IgG, and peptide microarray (Phadia) was used to detect antibodies against 10 citrullinated (Cit) peptidic antigens: CCP-1 (Filaggrin), CEP-1 (α-enolase), Vim 2-17, Vim 60-75 (Vimentin), Fib α 36-50, Fib α 573, Fib α 591, Fib α 621-635, Fib β 36-52, Fib β 60-74 (Fibrinogen). Most of the patients (n=93) were followed up after 6 months. Logistic regression analysis

was performed to examine associations between HRCT lung changes at the time of RA diagnosis and autoantibodies.

Results

HRCT parenchymal and airway changes was present in 58 (54.7%) and 68 (64.2%) patients, respectively. The forced vital capacity (FVC) was significantly lower in the presence of airway abnormalities, while the ratio FEV1/FVC was significantly lower in patients with parenchymal lung changes. Higher age, RF IgA, CCP2 IgG, ever smoking and pack-years above 24 were significant predictors of parenchymal lung changes. Some ACPA fine specificities, especially against Cit Fib and Vim peptides, were associated to parenchymal lung changes in ever smokers. The risk of having parenchymal changes increased parallel to the increase in number of ACPA specificities. Having more than 5 ACPA specificities at the time of diagnosis increased the risk of having parenchymal lung abnormalities in current smokers (OR=13.8, 95% CI=1.0-196.2, p=0.05). Of the patients that were followed up after 6 months 4 had progression of fibrosis and 3 had new fibrosis. No difference in airway changes were observed at follow-up. There was a significant decrease in DAS28 at follow-up (Mean±SD: 5.5 ± 1.1 vs 3.2±1.3 P value < 0.001). The titers of some but not all of the ACPA fine specificity was significantly decreased after 6 months of treatment CCP-1 (Mean±SD 46.6±289.7 vs 101.6±235.3), CEP-1 (59.1±120.1 vs 35.9±76.3), Vim 60-75 (338±488.1 vs 220.3±327.2), Vim 2-17 (25.7±59.3 vs 17±37.8), Fib β 60-74 (237.3±411 vs 136.4±241.5), Fib α 621-635 (187.3±306.3 vs 115.9±203.2).

Conclusions:

The presence of RF IgA, anti-CCP2 IgG and antibodies to Cit Fib and Vim peptides were associated with parenchymal lung changes in early-untreated RA. The more ACPA fine specificities, the higher the risk of having parenchymal lung changes already at the time of RA diagnosis. Treatment with DMARDs significantly reduces the disease activity and titers of some of the ACPA fine specificities.

5. Season of Birth and Cigarette Smoking as Modifiers of the Effect of HLA DRB1 Shared

Epitope on the Development of ACPA Positive RA. Results from a Swedish Population

Based Case-Control Study.

Patrik Stolt¹, Lars Klareskog², Leonid Padyukov², Camilla Bengtsson¹, Lars Alfredsson¹

1: Institute of Environmental Medicine, Unit of Cardiovascular Epidemiology, Karolinska Institutet,

171 77 Stockholm, Sweden. 2: Department of Medicine, Rheumatology Unit, Center for Molecular

Medicine, Karolinska University Hospital Solna, 171 76 Stockholm, Sweden

Abstract

Background: Shared epitope (SE) alleles, cigarette smoking and antibodies to citrullinated peptide

antigens (ACPA) are well recognized components in the development of Rheumatoid Arthritis (RA) (1).

More recently, the effect of early life exposures on this process has become a topic of interest (2). The

objective of this study was to analyse the combined effect of season of birth, used as a marker of

prenatal to early childhood exposures, SE alleles and cigarette smoking on the development of RA of

different ACPA phenotype.

Method: In a Swedish study of RA, 1726 cases and 3683 controls included October 2005 – September

2014 were investigated by analysing the effect of different combinations of season of birth, SE

genotype and cigarette smoking status on the risk of RA of different ACPA phenotype. The analyses

were based on comparisons of single SE (SSE) carriers and double SE (DSE) carriers with SE-non

carriers, of ever-smokers with never-smokers and of persons having different season of birth. Risk was

estimated by calculating odds ratio (OR) with 95% confidence interval (95% CI).

Results: Comparing persons having different month of birth, a tendency towards a comparably lower

risk of ACPA positive RA among persons born in July was observed, however inconclusively due to

wide and overlapping CI:s. Adding SE genotype as explanatory variable by analysing the variation of

the effect of carrying DSE, compared with not carrying SE, over different seasons of birth, a lower risk

estimate of ACPA positive RA was observed among persons born in the period of June – August than

among persons born in the other quarters of the year. Repeating these analyses among the ever-

smokers separately, a clearly lower risk estimate of ACPA positive RA was observed among persons

born in the period of June-August, the OR = 2.81, 95% CI 1.16-6.77, than among persons born at any

time of the year except June – August, the OR = 21.02, 95% CI 11.00-40.15, or at any time of the year,

OR = 11.44, 95% CI 6.96-18.82. An analogous variation was not observed among the never-smokers,

the corresponding OR being 7.12, 95% CI 2.33-21.76, 7.62, 95% CI 4.10-14.17 and 7.54, 95% CI 4.39-

12.96, respectively. The corresponding analyses in which SSE replaced DSE remained inconclusive due

to overlapping CI:s.

Conclusion: These results indicate that the effect of SE on the development of ACPA positive RA may

be modified by season of birth when combined with cigarette smoking in adulthood. This is suggestive

of some seasonal factor in the prenatal to early childhood period modifying the risk of developing RA

by interacting with gene polymorphisms and exposures later in life.

References:

1. Klareskog L et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger

HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by

citrullination. Arthritis Rheum. 2006;54:38-46.

2. Disanto G et al. Month of birth, vitamin D and risk of immune- mediated disease: a case

control study. BMC Medicine 2012;10:69.

6. Cloning of Gingival Tissue B Cells From an ACPA+ RA Patient with Periodontitis

Natalia Sherina1, Khaled Amara1, Natalie Sippl1, Daniel Ramsköld1, Johanna Steen1, Lena Israelsson1, Monika Hansson1, Annika van Vollenhoven1, Kaja Eriksson2, Tulay Yucel-Lindberg2, Vivianne Malmström1, Karin Lundberg1

1. Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Solna, Stockholm, Sweden

2. Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden

Background and objectives

Rheumatoid arthritis (RA) is characterized by autoantibodies to citrullinated proteins (ACPA). Recently Porphyromonas gingivalis, a major cause of periodontitis (PD), has been linked to ACPA+ RA. Our ambition with this study is to clone ACPA-specific B cells from gingival tissue (GT) of patients suffering from both PD and RA, in order to demonstrate that citrulline-specific B cells, previously only detected in RA joints and circulation, may also reside in gingival tissue.

Materials and Methods

Gingival tissue-derived single CD19+ B cells from an ACPA+ RA patient with PD (RA/GT) were sorted by flow cytometry. Immunoglobulin (Ig) variable region genes were sequenced and expressed to generate recombinant monoclonal antibodies (mAbs).

Results

We have isolated 480 CD19+ B cells from the gingival tissue, and analyzed 110 variable heavy chain Ig genes (IGHV). Ig gene sequence analysis demonstrated that the B cell repertoire was predominantly polyclonal, although two clonally related B cell populations (approximately 2%) were detected. Compared to B cells from peripheral blood of healthy controls, RA/GT B cells showed decrease in usage of VH3 and increase in VH4. By individual VH gene segments, IGHV4–31 was overrepresented compared to controls. Conversely, IGHV1-2, IGHV3-23, and IGHV4-34 were under-represented. Interestingly, antibodies with positively charged IGHV CDR3 regions, a feature associated with autoreactivity, were enriched in GT. By contrast, antibodies with negatively charged CDR3 regions were enriched in controls. Alignment of VH sequences to their closest germline counterparts revealed that RA/GT B cells exhibited extensive mutations in the IGH CDR regions and higher levels of somatic mutations in the V gene segments compared to controls, suggesting an antigen-driven B cell response. We have so far expressed 60 mAbs that we are currently screening for reactivity with citrullinated epitopes and bacterial antigens.

Conclusions

We have been able to successfully isolate and clone a number of gingival tissue-derived B cells. Based on the hypothesis that the ACPA-response may be initiated at mucosal surfaces such as gingival tissue, we now have the tools available to more directly address this etiological question.

7. A novel candidate for genetic control of Collagen Induced Arthritis is involved in transcriptional regulation of B-cell proliferation Samra Sardara, Alish Kerra,b, Daniëlle Vaartjesa,c, Mathilde Emilie Voetmanna,d, Emilie Riis Moltveda,e,

Åsa Anderssona,f

a Section for Molecular and Cellular Pharmacology, Department of Drug Design and Pharmacology,

University of Copenhagen, Copenhagen, Denmark

b Present address: Pfizer pharmaceuticals, Dublin, Ireland

c Present address: Division of Medical Inflammation Research, Department of Medical Biochemistry

and Biophysics, Karolinska Institutet, Stockholm, Sweden

d Present address: Biogen Denmark A/S, Hillerød, Denmark

e Present address: QuintilesIMS, North Carolina, United States of America

f School of Business, Engineering, and Science, Halmstad University, Halmstad, Sweden

Introduction: Rheumatoid Arthritis (RA) is a common autoimmune disease, caused by a complex

interplay of genetic and environmental factors. This project aims to investigate the role for the

transcriptional regulator TBX3 in the development of experimental arthritis. TBX3 was identified in

functional genetic studies of Collagen Induced Arthritis (CIA), an experimental model for RA.

Methods: CIA is induced in mice by immunization with collagen type II. The studies were performed

in mice congenic for a 1.4 Mbp genetic fragment on mouse chromosome 5 (Eae39r2). The whole

genomes of the parental mouse strains were sequenced by NGS. Congenic and control mice were

characterized by flow cytometry, in vitro lymphocyte activation assays, and transcript level studies in

order to identify the candidate gene. A time-course CIA experiment was performed to study protein

expression kinetics in relation to arthritis. CRISPR/Cas9 knock-out of the candidate gene in murine B

lymphocytes is currently underway.

Results: CIA experiments in Eae39r congenic, sub-congenic, and control mice have shown that a 1.4

Mbp sub-locus, within the Eae39r fragment, is controlling the severity of arthritis, in addition to anti-

collagen antibody titers. Within this sub-locus, a candidate gene, Tbx3, has been identified based on

its: (-) differential expression in the spleens and specifically in B cells of naïve congenic and control

mice; (-) various regulatory variations identified by NGS; (-) genetic conservation between mouse and

human; (-) a documented role in bone signaling pathways. In vitro lymphocyte activation and protein

activity experiments have demonstrated that the activity of TBX3 decreases in primary B-lymphocytes

upon stimulation through the B cell receptor. The protein activity also varies in B-lymphocytes at

different stages of CIA, hinting towards the importance in disease pathogenesis/progression.

Conclusion: We suggest that TBX3 is a novel candidate contributing to arthritis development by

controlling B lymphocyte activation. Studies of associated pathways will provide new insights into

disease pathogenesis and potential drug targets.

8. Identifying novel drug targets of inflammation by customized CRISPR/Cas9 approach

Authors: Sudeepta Kumar Panda, Zsolt Kasza, Sanjaykumar Boddul, Yanek Jiménez, Gustaf

Wigerblad, Long Jiang and Fredrik Wermeling

Affiliations: SKP, ZK, SB, YJ, GW, LJ, FW: Rheumatology Unit, Department of Medicine Solna,

Karolinska Institutet, Stockholm, Sweden.

Background: - Inflammation is a protective body response towards foreign invaders and ultimately

helps to restore body homeostasis. However, excessive and misdirected inflammation can result in

allergy and autoimmune disorders. Therefore, there is a huge demand to discover potent anti-

inflammatory drugs. In the clinic, high dose intravenous immunoglobulin (IVIG) administration is

used in severe inflammatory diseases such as Idiopathic thrombocytopenic purpura, and Kawasaki

disease, but also occasionally in Myositis and Rheumatoid Arthritis. On note, effectiveness of IVIG

depends on the minute expression of the IL-4Rα in myeloid effector cells. Interestingly, mice injected

with any inflammatory stimuli results in myeloid IL-4Rα expression, mediated by a soluble proteinase-

k sensitive factor that is secreted from bone marrow-derived cells found in fat pads and lung

(Wermeling et al., PNAS 2013). That we have referred to as the IL-4Rα regulating factor (IRF). In

general, neutrophils have low IL-4Rα expression, whereas during inflammation there is a massive

upregulation of this receptor, which has been a great paradox for us.

Aim: - Here we aim to investigate this soluble factor, possibly a novel biomarker of inflammation, in

myeloid cells using a CRISPR/Cas9 screening approach.

Methods and Results: - First, we established an in vitro model system in which in which

supernatant from LPS challenged mouse alveolar macrophages (MH-S) and conditional Hoxb8

macrophage cell line selectively upregulated IL-4Rα expression in bone marrow-derived neutrophils.

Secondly, different MH-S clones (good and poor IRF producers) were challenged by LPS and gene

expression was determined by microarray. Thirty-four different candidate genes for the IRF were

identified comparing the good and poor IRF producers with control. Among 34 candidates list, seven

top most genes that are highly upregulated IL-4Rα expression in good clones were used in the CRISPR

screen purpose. To simplify the generation of such custom CRISPR screens we have furthermore

developed a software called Green Listed (Panda et al., Bioinformatics 2016). Thirdly, we established a

stable Cas9 expressing Hoxb8 macrophage cell line (Hoxb8 macrophageCas9) that serve as a host for a

lenti viral guide RNA library. This Hoxb8 macrophageCas9 cells were infected with the lenti viral library

at a very low MOI (MOI < 0.03) to integrate one guide RNA per one cell. To this, we established 132

different single cell clones. Consecutively, we repeated the in vitro bone marrow assay in neutrophils

by priming the supernatant of these LPS challenged clones.

Conclusion: We are currently screening these clones to identify the gene, as an IL-4Rα regulating

factor (IRF), responsible for the myeloid IL-4Rα expression. This study will delineate the novel

regulatory pathways that targets STAT6 signaling to myeloid cells that could have implications for

several inflammatory and autoimmune diseases.

9. Machine learning using rheumiatoid arthritis synovial gene expression data defines

histologic features that correlate with systemic inflammation and autoantibodies

Dana Orange

Rockefeller University

Objective: We sought to refine histologic scoring of rheumatoid arthritis synovial tissue by training

scoring metrics with transcriptome-wide expression data and machine learning.

Methods: Twenty histologic features were assessed on 140 synovial tissue samples. Consensus

clustering of gene expression data from a subset of 45 synovial samples identified three unbiased and

robust subtypes. Support vector machine (SVM) learning was then used to predict gene expression

subtypes using histology data as the descriptive features. Corresponding clinical and laboratory data

were compared across subtypes.

Results: Consensus clustering of RNA-seq data revealed three distinct synovial subtypes of RA,

including [i] a highly inflammatory subtype characterized by extensive infiltration of leukocytes, [ii] a

low-inflammatory, or fibroid subtype characterized by enrichment in pathways including TGF-β,

neuronal genes and mucin deposition, and [iii] a mixed subtype. Patients with highly inflammatory

synovial subtypes exhibited higher levels of markers of systemic inflammation and autoantibodies, but

not increased joint counts or pain relative to other subtypes. Though CRP and ESR correlated with

pain in the highly inflammatory patients, there was no evident relationship for the fibroid samples.

Machine learning algorithms applied to histology features using RNA-seq subtypes as data labels

yielded good predictive scores, particularly for the high and low inflammatory subtypes, revealing good

synchrony between RNA-seq data and the histology features selected for this study.

Conclusion: Careful analysis of RNAseq data revealed three distinct synovial subtypes that associate

with levels of ESR, CRP and autoantibodies, and a learning algorithm using histology scores and

subtype labels distinguished the subtypes successfully. Comparison of gene expression patterns to

clinical features revealed a potentially clinically important distinction: mechanisms of pain may differ

in patients with high ESR and a synovial inflammatory expression pattern from those with low ESR

and a fibroid synovial expression pattern.

10. NEW MUTATED PEPTIDYLARGININE DEIMINASE FROM PORPHYROMONAS GINGIVALIS A TARGET IN EARLY RA CITRULLINATES MAJOR RA-AUTOANTIGENS

M. Jenning1, B. Marklein1, J. Yetterberg2, G. Burmester1, K. Skriner1,*

1Department of Rheumatology and Clinical Immunology, CHARITÉ UNIVERSITY MEDICINE, Berlin, 2Department of Rheumatology and Clinical Immunology, Karolinska Institutet, Stockholm, Germany

Objectives: Previous reports showed that peptidylarginine deiminase (PPAD) form Porphyromonas gingivalis (P.g.) is not able to citrullinate proteins internaly. New mutated PPAD (mPPAD) from P.g. involved in periodontal disease (PD) cloned out of P.g. strain was characterized and analyzed for its reactivity in sera from patients with systemic autoimmune diseases

Methods: We cloned a new enzymatically active recombinant mutated PPAD from P.g. mPPAD mutations and citrullination sites were analyzed by DNA sequencing and/or protein mass spectrometry. Autocitrullination activity it´s enzymatic-activity and human autoantigen protein citrullination was investigated by 2D-Elektrophoresis, MS, immunoblot analysis and ELISA. Furthermore we tested anti-mPPAD/cit-mPPAD with human sera (n=93) from early RA before and after onset of RA (n=30), established RA (n=32), SLE (n=16) and healthy blood donors (n=15) in ELISA assays. To study a potential impact on the RA mouse model (CAIA), mPPAD-containing vesicles from P.g. were injected by intraperitoneal injection (IP).

Results: Recombinant mPPAD lacks 43 amino acids at the N-terminus and exhibits so far two new amino acid mutations (amino acid position 73 (F>L) and 447 (E>V). We were able to demonstrate, mPPAD is enzymatically active over a huge pH-range (3-10) and autocitrullinates at amino acid position 63 the arginine to citrulline. Moreover mPPAd citrullinates major autoantigens in RA (Fibrinogen, Vimentin and hnRNP-A2/B1) which are detectable by RA patient sera and specific anti-citrulline monoclonal antibodies. mPPAD citrullinates HeLa-protein extracts and these specific citrullinated proteins are recognized by RA patient sera. Anti-citrullinated mPPAD antibodies were detected in 41% (n=32) of patients with RA but not in SLE (n=16) and control sera (n=15). In a RA follow-up study (n=30), we detected nearly similar antibody-sensitivities for citrullinated mPPAD before and after onset of RA (13/20%). Only a minority (7%) of RA patients show higher mPPAD antibody levels after RA diagnosis. In the Collagen antibody-induced arthritis (CAIA) RA mouse model mPPAD containing P.g. vesicles when injected IP showed a TLR2-dependent protective anti- inflammatory effect like P.g. LPS and Lipomannan.

Conclusions: Pg. infection and RA disease diagnosis occurs on different timepoints and Pg. infection induces a TLR2-dependent protective anti-inflammatory effect.We show the first time that mPPAD can citrullinate major human autoantigens internally and their immunologically and diagnostic relevance.

11. A long non-coding RNA in the rheumatoid arthritis risk locus at chromosome 18 is involved in T cell activities M. Houtman, K. Shchetynsky, L. Padyukov. Rheumatology Unit, Department of Medicine Solna, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden. Rheumatoid arthritis (RA) is a relatively common autoimmune disorder that is characterized by joint inflammation and destruction. Genome-wide association studies have enabled the discovery of common genetic variations in over 100 loci contributing to RA, but the precise targets of the majority of these associations are unknown. We aimed to identify the molecular mechanisms that connect variations in the protein tyrosine phosphatase non-receptor type 2 (PTPN2) locus with RA. We have found that previously identified RA-susceptibility variants in the PTPN2 locus are associated with the expression of a long non-coding RNA LINC01882 located 30kb downstream of PTPN2 (whole blood, GTEx). To identify the function of this long non-coding RNA, we suppressed the expression of LINC01882 using antisense oligonucleotides and RNA interference in Jurkat T cells. Twenty-four and 48 hours after knockdown, total RNA was extracted and ribosomal RNA was depleted. Sequencing libraries were generated using the Illumina TruSeq Stranded Total RNA kit and sequenced on an Illumina HiSeq 2500 platform, yielding about 25 million paired-end reads per sample. Differential expression analyses were conducted using DESeq2. We identified 12 deregulated genes after LINC01882 knockdown (6 upregulated and 6 downregulated; FDR < 0.05). One of the downregulated genes is the zinc finger E-box-binding protein 1 (ZEB1), a repressor of interleukin-2 (IL-2) gene transcription. This points towards the involvement of LINC01882 in T cell differentiation. Our data suggest that LINC01882 might contribute to the molecular mechanism underlying RA. Overall, these findings suggest that previous annotation of the associated locus to PTPN2 is potentially incorrect and that the long non-coding RNA LINC01882 might be the causal gene. Overall, these findings point towards the potential role of LINC01882 in the RA susceptibility association attributed to the PTPN2 locus.

12. Elevated serum IL-25 in rheumatoid arthritis patients with bone erosion and

interstitial lung disease

LU Jinyue1,DA Minglian1,FENG Yuchen1, LIU Yuanyuan2,ZHANG Sigong2,SHEN Haili*

1Second Clinical Medical College, Lanzhou University ,Lanzhou 730000;2Department of

Rheumatology, Lanzhou University Second hospital, Lanzhou 730000,China

*Corresponding author, E-mail:[email protected]

Objective To detect the serum levels of IL-25 and IL-17 in rheumatoid arthritis (RA) patients and

investigate the potential relationship with bone erosion and concomitant interstitial lung disease

(ILD). Methods The study enrolled a total of 117 RA patients and 56 healthy subjects as control. The

serum levels of IL-25 and IL-17 were determined by ELISA, and RF was detected by turbidimetric

immunoassay, anti-CCP antibody as well as erythrocyte sedimentation rate (ESR) and C-reactive

protein (CRP)were also tested. ILD was identified on high-resolution computed tomography (HR-CT),

the degree of bone erosion was inspected by musculoskeletal ultrasoum examination, and radiographic

grade was graded by Sharp-van der Heijde Score (SHS), Disease activity in RA was scored with the

DAS28 and visual analogue scale(VAS). correlation analysis was used to evaluate the correlations of

IL-25 and IL-17 in different groups. Results Compared with heathy control group, the serum levels

of IL-25 and IL-17 were increased significantly in patients with RA(graph A,B). Compared with bone

erosion negative group, the serum level of IL-25 was higher significantly in bone erosion group(graph

C,D). The level of IL-25 was higher in the ILD group of RA patients than the non-ILD group(graph

E,F). In addition, there were positive correlation between the serum level of IL-25 and RF-

IgG(r=0.367), RF-IgA(r=0.319), RF-IgM(r=0.436)( graph G). Meanwhile, the serum level of IL-17 had

the positive correlation with RF-IgG(r=0.216), RF-IgM(r=0.271) . both of them have no correlation

with anti-CCP antibody. Conclusion The serum level of IL-25 is raised in RA patients with bone

erosion and ILD.

[Key words] rheumatoid arthtitis;cytokines;bone erosion;intersititial lung disease ;inflammatory

13. ALTERED LYMPH NODE STROMAL CELLS DURING THE EARLIEST PHASES OF RHEUMATOID ARTHRITIS

C. Ospelt1, E. Karouzakis1, J. Hähnlein2, J.F. Semmelink2, R.E. Gay1, P.P. Tak2, 3, D.M. Gerlag2, 4, S. Gay1, L.G.M. van Baarsen2

1Center of Experimental Rheumatology, University Hospital of Zurich, Zurich, Switzerland, 2Academic Medical Center, Amsterdam, Netherlands, 3Cambridge and GlaxoSmithKline, University of Cambridge, Stevenage, 4R&D Projects Clinical Platforms & Sciences, GSK Clinical Unit Cambridge, Cambridge, United Kingdom

Background: Rheumatoid arthritis (RA) is an autoimmune disease with unknown etiopathogenesis where systemic autoimmunity precedes clinical disease onset. Adaptive immunity is initiated in lymphoid tissue where lymph node stromal cells (LNSC) play a crucial role in shaping the immune response and maintaining peripheral tolerance. We developed an experimental model for studying the functional capacities of human LNSC during the earliest phases of RA and compared their cellular and molecular characteristics to LNSC from healthy volunteers.

Methods: ACPA+ RA patients (n=24), ACPA+ RA-risk individuals (n=23) and seronegative healthy controls (n=14;HC) underwent ultrasound-guided inguinal lymph node biopsy. Human LNSCs were isolated and expanded in vitro for cellular (flow cytometry), molecular (methylome, transcriptome and microRNA) and functional analyses.

Results: Key LN chemokines CCL19, CCL21 and CXCL13 were induced in LNSCs upon stimulation with TNFα and lymphotoxin α1β2, but to a lesser extent in LNSCs from RA patients. RNA sequencing was performed on LNSC of HC (n=5), ACPA+ RA-risk individuals (n=6) and ACPA+ RA patients (n=4). Of interest, LNSC from ACPA+ RA-risk individuals and ACPA+ RA patients revealed a common significantly differential expressed gene signature compared with HC LNSC. Pathway analysis of this common signature showed, among others, significant enrichment of pathways affecting actin cytoskeleton, focal adhesion and cell junction. Accordingly, in a gel contraction assay LNSC from ACPA+ RA-risk individuals and RA patients showed impaired collagen contraction compared to healthy LNSC. In RA LNSC a significant enrichment was observed for genes involved in TGFb signalling while in RA-risk LNSC cell cycle genes were differentially expressed compared with HC. DNA methylation analyses revealed common differentially methylated CpG sites (DMS) in LNSC from ACPA+ RA patients (n=5) and ACPA+ RA-risk individuals (n=3) compared with HC (n=4). These DMS were significantly hypomethylated and associated with antigen processing and presentation (HLA-DRB1).

Conclusions: This data point towards alterations in the cytoskeleton and antigen-processing and presentation in LNSC from ACPA+ RA-risk individuals and RA patients. Further studies are required to investigate the influence of this LNSC abnormality on immune responses.

14. Receptor activator of nuclear factor kappa-B ligand (RANKL) and

Marginal jawbone loss predates the onset of rheumatoid arthritis.

Johansson L3, Kindstedt E1, Palmqvist P1, Koskinen-Holm C1, Kokkonen H3, Johansson I2, Rantapää

Dahlqvist S3*, Lundberg P1*

INTRODUCTION: Previous studies have shown a higher incidence of alveolar bone loss in patients with rheumatoid arthritis (RA) and that patients with periodontitis are at a greater risk for developing RA. Periodontitis, displayed as marginal jawbone loss was analysed in individuals prior to symptom onset of RA and related to plasma levels of receptor activator of nuclear factor kappa-B (RANKL), a cytokine crucial for bone resorption.

METHODS: A case-control study performed within the Medical Biobank of Northern Sweden included 232 pre-symptomatic individuals with blood samples donated before symptom onset and 194 controls. A questionnaire on self-assed dental status and smoking status was retrieved. Dental radiographs to evaluate marginal jawbone levels were available from 93 pre-symptomatic individuals (mean age; 56.8 95%CI55.9, 57.7 years and pre-dating time; -5.3 95%CI -12.2, -0.2, 74.2% females) and 83 controls (mean age; 55.5 95%CI54.6, 56.5, 73.5% females) . Of these individuals 45 had radiograph documentations prior to development of RA symptoms and to whom sex, age and smoking status could be matched among the controls. Plasma were analysed for RANKL (BioVendor, Karasek, Czech Republic), and anti-citrullinated peptide antibodies (ACPA) (anti-CCP2 test, Eurodiagnostics, Sweden) from similar time points.

RESULTS: Compared to matched controls, total bone loss was significantly higher in never-smokers who developed RA but not in smokers and increasing levels on total jawbone loss was associated with a significantly higher odds to be diagnosed with RA later (OR=1.06, 95%CI 1.01, 1.11). Regardless of smoking status, the number of unaffected teeth did not differ significantly between those who were subsequently diagnosed with RA and their matched controls. In the pre-symptomatic individuals RANKL positive individuals had significantly higher extent of marginal jawbone loss, which was further increased in ACPA positive individuals. Previously documented association between smoking and ageing and marginal jawbone loss was verified.

CONCLUSIONS: Marginal jawbone loss preceded onset of symptoms of RA but the difference was only manifested in non-smokers. Moreover, marginal jawbone loss and plasma RANKL levels were related in the pre-symptomatic individuals particularly in ACPA positive individuals.

15. The association between HLA-DRB1 alleles and risk of rheumatoid arthritis is

influenced by massive gene-gene interactions.

Lina-Marcela Diaz-Gallo,1* Daniel Ramsköld,1,7 Lasse Folkersen,2,7 Klementy Shchetynsky,1,7 Karine

Chemin,1 Boel Brynedal,3 Steffen Uebe,4 Yukinori Okada,5,6 Lars Alfredsson,3 Lars Klareskog,1 Leonid

Padyukov1**

1. Rheumatology Unit, Department of Medicine Solna, Karolinska Institutet, Karolinska

University Hospital, Stockholm, 171 76 Sweden.

2. Department of Bioinformatics, Technical University of Denmark, DK-2800 Lyngby,

Denmark

3. Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.

4. Human Genetics Institute, Universitätsklinikum Erlangen, Erlangren, Germany.

5. Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital.

Harvard Medical School, Boston, Massachusetts, USA.

6. Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-

IFReC), Osaka University, Suita, Japan.

Correspondence: * [email protected], @LinMarDiaz ** [email protected]

In rheumatoid arthritis (RA) a particular subset of HLA-DRB1 gene variants provide a higher risk

factor. A common way of describing these subsets has been by naming them the “shared epitope (SE)”

refereeing to amino acid sequences known to be coded by the susceptibility gene variants. Here, we

aimed to investigate how gene-gene interactions influence this major HLA-DRB1 related disease risk

and more specifically we set out to investigate whether a large number of non-HLA SNPs with

individually low risk to develop RA modulate the HLA-DRB1 effect to develop RA.

In order to address this question, we measured additive interaction at GWAS level for two independent

RA cohorts, the Swedish EIRA and the North American NARAC. We computed the attributable

proportion (AP) due to interaction between HLA-DRB1 SE and a set of SNPs associated with disease or

not-associated with disease in these cohorts (in total more than 5 million SNPs). The distributions of

the observed AP p-values were compared by the Kolmogorov-Smirnov test. We found a strong

enrichment of significant interactions (AP p-value <0.05) between HLA-DRB1 SE alleles and a group

of SNPs associated with RA in both cohorts, evidenced by the distribution test (KS-test D=0.35 for

EIRA and D=0.25 for NARAC, p<2.2e-16 for both).

Our data indicate that the strong association between HLA-DRB1 SE alleles and risk of anti-

citrullinated protein antibody (ACPA) positive RA is modulated in a major way by massive interactions

with non-HLA genetic variants.

16. Characterisation of the antibody response to a citrullinated peptide derived from Porphyromonas gingivalis PAD in RA Nastya Kharlamova1, Boel Brynedal2, Xia Jiang2, Natalia Sherina1, Kaja Eriksson3, Tulay Lindberg3, Monika Hansson1, Lena Israelsson1, Johanna Steen1, Vivianne Malmström1, Lars Alfredsson2, Khaled Amara1, Karin Lundberg1

1 Rheumatology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden 2 Cardiovascular Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden 3 Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden

Objectives: Anti-citrullinated protein antibodies (ACPA) - a hallmark of rheumatoid arthritis - can be detected years before clinical signs of joint inflammation. Since citrullination and expression of peptidyl arginine deiminase (PAD) (i.e. the enzyme responsible for citrullination) is an inflammation-dependent process, and generally not present in healthy joints, it has been suggested that break of immune tolerance to citrullinated proteins occurs at an extra-articular site. The possible involvement of the gum mucosa has been discussed, primarily based on the unique feature of Porphyromonas gingivalis (P.g) - a keystone pathogen in periodontitis (PD) - to express a bacterial version of the PAD enzyme (denoted P.PAD), capable of autocitrullination. In the present study, we have investigated the antibody response against CPP3, a citrullinated peptide derived from P.PAD, in order to address the hypothesis that P.g may drive ACPA-production. Methods: This study includes: 2,859 RA cases and 4,864 controls from the Epidemiological Investigation of RA (EIRA) cohort; 65 PD patients and 59 periodontally healthy individuals; and 218 monoclonal antibodies, derived from RA B cells. Reactivity with citrullinated P.g and human antigens was assayed by ELISA and/or multiplex. Associations with genetic risk factors and smoking were determined by logistic regression. ACPA co-occurrence was analysed by pairwise Pearson correlation. Monoclonal antibody mutations were analysed by IgBLAST comparison. Results: Anti-CPP3 IgG was detected in 11% of RA, 10% of PD and <2% of controls, with higher levels in RA compared to PD. These antibodies clustered outside the classical ACPA response, associated with smoking, but not with major genetic risk factors. Two CPP3-reactive monoclonal antibodies were identified; one which cross-reacted with citrullinated human vimentin and had extensive mutations, indicating antigen-driven clonal selection and affinity maturation. Conclusion: Based on our data, we propose that P.g infection triggers an antibody response to CPP3, which cross-reacts with citrullinated human proteins by mechanisms of molecular mimicry. Future studies should address whether anti-CPP3 IgG could serve as a biomarker to identify individuals with PD at increased risk for RA.

17. PTPN22 - novel gene to function data for T cells in Rheumatoid Arthritis

Karine Chemin1, Daniel Ramsköld1, Lina-Marcela Diaz-Gallo1, Jessica Herrath1, Karolina Tandre2, Lars Rönnblom2, Anca Catrina1, Vivianne Malmström1

The presence of the PTPN22 risk allele (1858T) is associated with several autoimmune diseases including rheumatoid arthritis (RA). Despite a number of studies exploring the function of PTPN22 in T cells, the exact impact of the PTPN22 risk allele on T-cell function in humans is still unclear. In this study, using RNA sequencing, we show that, upon TCR-activation, naïve CD4+ T cells homozygous for the PTPN22 risk allele overexpress a limited number of genes including CFLAR and 4-1BB, which are important for cytotoxic T-cell differentiation. Moreover, the protein expression of the T-box transcription factor Eomesodermin (EOMES) was increased in T cells from healthy donors homozygous for the PTPN22 risk allele. No difference in the frequency of other CD4+ T cell subsets (Th1, Th17, Tfh, Treg) was observed in healthy donors homozygous for the PTPN22 risk allele and suppressive capacity of Treg cells was not altered. Finally, an accumulation of EOMES+CD4+ T cells was observed in synovial fluid of RA patients with a more pronounced production of Perforin-1 in PTPN22 risk allele carriers. Altogether, we propose a novel mechanism of action of PTPN22 risk allele through the generation of cytotoxic CD4+ T cells and identify EOMES+ CD4+ T cells as a relevant T-cell subset in RA pathogenesis.

1Rheumatology Unit, Department of Medicine, Karolinska University Hospital Solna, Karolinska Institute, Stockholm, Sweden

2Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden

18. Monoclonal ACPA-IgG feature extensive Fab glycosylation

Katy A Lloyd1, Johanna Steen1, Phillip J Titcombe1,2, Khaled Amara1, Diana Zhou1, Lena Israelsson1, Susanna L Lundström1,3, Daniel L Mueller2,4, Lars Klareskog1, Vivianne Malmström1, Caroline Grönwall1

1Department of Medicine, Rheumatology Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; 2Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA; 3The Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.

4. Division of Physiological Chemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden

Background

Fab-glycosylation is found in ~15-25% serum IgG and while its exact consequence remains unknown, it may alter IgG functionality. Recent data revealed elevated Fab-glycosylation in polyclonal anti-citrullinated protein autoantibodies (ACPA) from rheumatoid arthritis (RA) patients. Herein, we characterize the Fab-glycan profile of monoclonal ACPA.

Methods

A total of 14 recombinant human ACPA, derived from RA synovial plasma cells or circulating memory, were evaluated for predicted N-linked glycosylation sites using the NetNGlyc server. Fab-glycosylation was verified with enzymatic digestion, Western blot, lectin-ELISA, and mass spectrometry. Antigen binding was investigated by CCP3 ELISA. VH-VL structure models were generated using the PIGS tool and the GlyProt server. The frequency of predicted VH-VL sites was compared to single-cell paired heavy and light chains from extensively mutated mAbs (>15 mutation in VH or VL): 51 expressed non-ACPA synovial B cells from seropos. RA, and 27 from seroneg. RA, and 198 bone marrow (BM) plasma cells, and 27 clones from healthy control circulating memory. These were compared to 19 highly-mutated broadly-neutralizing (bn) HIV mAbs and 103 plasmodium faciparium (PF) specific mAbs from the literature. Fisher’s exact test or Kruskal-Wallis test was used in statistical analysis.

Results

The majority of ACPA exhibited variable region N-linked motifs (85.7%), compared to 18.5% in control (p<0.0001), 21.2% in RA BM plasma cells (p<0.0001), 31.4% non-ACPA synovial RA mAbs (p=0.0005), 7.4% of clones from seroneg. RA (p<0.0001), 25.2% in PF mAbs (p<0.0001), and 63.2% of HIV bnAbs (p=0.24), featured in both framework and CDRs generated by somatic hypermutation (SHM). Indeed, ACPA displayed high level of SHM (average 30 VL and 52 VH), yet when adjusted for SHM, N-linked motifs were significantly elevated in ACPA compared to all groups including bnAbs. IgG mAb characterization revealed that N-linked motifs were indeed glycosylated, although preliminary data suggested that glycans had no striking effect on antigen-binding. Homology-based structures predicted glycans to be primarily positioned outside of the potential antigen-binding site. Lectin analysis and mass spectrometry suggested that ACPA mAb Fab-glycan composition was distinctly different from Fc-glycans, and could have high sialic acid content.

Conclusions

The results support that Fab glycosylation is a key feature of ACPA. Significant increases in N-linked motifs in ACPA compared to other highly-mutated antibodies signifies that this is not solely associated to mutation frequency. Future studies are merited to further investigate the selection mechanisms and functional role of Fab-glycosylated autoantibodies.

19. Autoantibodies to post-translational modifications in Rheumatoid Arthritis are highly diverse in characteristics and functionality.

Johanna Steen1, Peter Sahlström1, Victoria Odowd2, Akilan Krishnamurthy1, Lena Israelsson1, Monika Hansson1, Linda Mathsson Alm3, Karl Skriner4, Stephen Rapecki2, Holger Bang5, Anca Catrina1, Daniel Lightwood2, Lars Klareskog1, and Vivianne Malmström1

1 Karolinska Institutet, Stockholm, Sweden

2 UCB, Slough, England

3 Thermo Fisher, Uppsala, Sweden

4 Charité Univeristätsmedizin, Berlin, Germany

5 Orgentec Diagnostika, Mainz, Germany

Rheumatoid Arthritis (RA) is a common inflammatory autoimmune disease that is characterized by a sub-population harboring autoantibodies to citrullinated antigens (ACPA). Patients that are ACPA positive suffer from a more severe disease and this set of autoantibodies have been postulated to be involved in the RA pathogenesis. However, ACPA appear years before disease onset, and thus have this notion been questioned.

We have expressed monoclonal plasma cell derived IgG antibodies from an RA patient’s joint. We showed that ACPA were highly mutated as compared to the nonACPA antibodies and were not phylogenetically related, and thus originated from separate immune responses. One of the four identified ACPA were clonally expanded.

The four ACPAs were all highly cross-reactive with different citrullinated targets, both peptides and proteins, however did they never target any non-modified antigens. None of the ACPA had identical citrulline recognition pattern. One of the autoantibodies also detected carbamylated antigens, another post-translational modification related to RA autoimmunity.

We made chimeric antibodies where we removed the hypermutations in either the heavy or the light chain, to dissect their contribution. We concluded that mutations could be restricted to one of the chains (either the light or the heavy), with retained citulline reactivity.

To investigate if the ACPA could have effect on bone destruction in RA, we tested the ACPAs in in vitro assays. Two of the ACPA had effect – one had a promoting effect, and one had an inhibitory effect in vitro. This highlights the importance of monoclonal investigations of ACPA functions.

20. Occupational Exposure to Asbestos and Risk of Rheumatoid Arthritis Authors: Anna Ilar1, Per Gustavsson1,2, Pernilla Wiebert1,2, Camilla Bengtsson1, Lars Klareskog3, Lars Alfredsson1,2 1. The Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden

2. Centre for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden 3. Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockholm, Sweden Background Due to the known association between silica dust and rheumatoid arthritis (RA), we wanted to study the association between RA and another silicate mineral; asbestos. The aim was to estimate the risk of seropositive or seronegative RA from ever occupational asbestos exposure as well as years with exposure. Methods The study base consisted of men and women living in Sweden between 2006 and 2013. RA patients were identified from the Swedish Rheumatology Quality Register (SRQ). We matched ten controls from the national population register per case on age, county and sex. Data on occupational histories were collected from the national population and housing censuses carried out in 1960, 1970, 1975, 1980 and 1990. A job-exposure matrix (JEM) containing historical exposure estimates from 1955-1995 to asbestos was applied to the study participants’ occupational histories. We used conditional logistic regression to assess the odds ratios (ORs) and 95 % confidence intervals (CIs) of RA associated with ever exposure and years of exposure to asbestos. ORs were adjusted for ever exposure to silica dust and household disposable income divided into quartiles. Results 9 704 cases and 90 271 controls were included in the analysis. Ever vs. never asbestos exposure resulted in an OR of 1.35 (95 % CI: 1.22-1.48) among men and 1.10 (95 % CI: 0.94-1.29) among women for seropositive RA. The ORs decreased to 1.12 (95 % CI: 1.01-1.25) and 1.02 (95 % CI: 0.86-1.22) for men and women respectively after adjusting for silica exposure and household disposable income. Asbestos exposed men were more likely than women to have worked with asbestos for a longer period of time and their risk of seropositive RA increased with years with the exposure. Male participants with more than 20 years of asbestos exposure at work had an adjusted OR of 1.27 (1.06-1.53, p for trend: 0.008). Conclusions Asbestos exposure is associated with seropositive RA among men. The increased risk remained after adjustments for potential confounding from silica exposure and household disposable income.

21. Assay comparison for measurement of drug level and immunogenicity in rheumatoid arthritis patients.

Christina Hermanrud1, Karen Hambardzumyan2, Malin Ryner1, Per Marits2, Saedis Saevarsdottir2 and Anna Fogdell-Hahn1.

1Clinical Neuroimmunology, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden. 2Rheumatology Unit, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.

Infliximab and adalimumab are tumor necrosis factor-alpha (TNF-α) inhibitors used for treatment of rheumatoid arthritis (RA). Low drug levels often correlate with anti-drug antibodies (ADA) and are

likely to result in poor clinical outcome. Serum anti-TNF-α drug levels and ADA are screened with an

in-house validated ELISA used in clinical routine at Karolinska University hospital. However, samples with a drug level above 0.2µg/mL cannot be screened for ADA due to the assay´s drug tolerance.

Moreover, immune complexes could occur, thus making ADA undetectable yielding false negative

results. We determined the correlation of drug levels and ADA between the ELISA with measurement

of neutralizing ADA (bioassay, iLite) and immune complexes (PandA with acid dissociation). For 84 RA serum samples, there were a 100% correlation of infliximab and adalimumab levels

between ELISA and iLite. All infliximab treated patients (n=17) with low drug levels

were identified as ADA positive with ELISA, but only seven were characterized with neutralizing ADA with iLite. This lack of neutralizing capacity was not in agreement with

the low drug level. RA patients treated with infliximab (n=101) were analyzed with ELISA and

PandA. After 21 months post treatment initiation, 28 patients had no measurable drug levels and 19 were ADA positive. Only 20 patients had optimal drug levels. Serum samples with a drug level above

0.2µg/mL were screened for ADA with PandA and up to 62% were positive. The clinical relevance of

screening for immunogenicity of drugs and potential possibilities to aide optimization of treatment will

be evaluated.

22. Risk Factors for Cardiovascular Disease Predate the Onset of Symptoms of Rheumatoid Arthritis

Heidi Kokkonen1, Hans Stenlund2, and Solbritt Rantapää Dahlqvist1

1Department of Public Health and Clinical Medicine, Rheumatology, Umeå University, Umeå, Sweden, 2Department of Public Health and Clinical Medicine, Epidemiology and Global Health, Umeå University, Umeå, Sweden

Background: Patients with rheumatoid arthritis (RA) are at increased risk of developing cardiovascular (CV) comorbidity compared with the general population. Contradictory results concerning CV disease prior to onset of RA have been reported. Of the known CVD risk factors, a more atherogenic lipid profile and smoking have been presented prior to RA onset.

In this study, lifestyle factors, lipid levels, presence of hypertension and diabetes were evaluated in individuals prior to onset of symptoms of RA and matched population controls from northern Sweden.

Methods: A nested case-control study was based on population surveys from The Västerbotten Intervention Programme (VIP) and the WHO Multinational Monitoring of Trends and Determinants in Cardiovascular Disease (MONICA). Data were collected by a questionnaire (socioeconomic and lifestyle factors), assessments by a nurse (body mass index; BMI, waist and blood pressure), and blood sampling. The registers of patients with RA (ARA criteria) attending the Department of Rheumatology, Umeå was co-analysed with the registers from VIP and MONICA. This study included 547 pre-symptomatic individuals (median age 50.2 years; 372f/175m, median (IQR) predating time 5.0 (7.0) years), and 1641 controls (median age 50.3 years; 1116f/525m). CVD risk factors were defined as: hypertension (systolic ≥140 mmHg and/or diastolic ≥90 mmHg including hypertensive treatment), elevated ApoB/ApoA1 ratio (females ≥0.7, males ≥0.8, including lipid lowering treatment), BMI ≥25, diabetes, and ever smoker.

Results: In conditional logistic regression models elevated ApoB/ApoA1 ratio (OR 1.3 (95% CI 1.0,1.6)), smoking (OR 2.0 (95% CI 1.6,2.5)), BMI≥ 25 (OR 1.3 (95% CI 1.1,1.6)) and diabetes (OR 2.0 (95% CI 1.1,3.7)) were associated with individuals who subsequently developed RA. In women elevated ApoB/ApoA1 (OR 1.4 (95% CI 1.1-1.8)), smoking (OR 1.9 (95% CI 1.1-1.8)), and BMI ≥25 (OR 1.3 (95% CI 1.1-1.7)) were significant for being pre-symptomatic for RA, in men the risk factors were smoking (OR 2.2 (95% CI 1.5-2.5), and diabetes (OR 4.7 (95% CI 1.71-13.1)). Stratifying on the median age, the factor remaining significant for the future RA group in older individuals was smoking (OR 1.7 (95% CI 1.2,2.5)), whereas in the individuals ≤50.2 years the factors were: elevated ApoB/ApoA1 ratio (OR 1.4 (95% CI 1.0,1.9)), BMI ≥25.0 (OR 1.5 (95% CI 1.0-2.0)), and smoking (OR 2.1 (95% CI 1.5,3.0). The pre-symptomatic individuals had significantly higher frequency of risk factors, 42% had ≥3 of these compared with 30% of the matched controls (OR 2.8 (95% CI 1.8, 4.5)). Especially, ACPA positive pre-symptomatic individuals had a high OR for future RA when having ≥3 of the CV risk factors (OR 5.7 (95% CI 2.1-15.3)).

Conclusion: Several of the CV risk factors were present in individuals already years before onset of symptoms of RA. One third of the pre-symptomatic individuals had at least 3 of these factors present. The risk factors for CVD associated with future RA differ between women and men. In younger individuals as well as ACPA positive individuals the CVD risk factors have a greater impact. These results urge an early CV prevention in patients with RA.

23. Autoreactivity to malondialdehyde-modifications in rheumatoid arthritis is linked to disease activity and synovial pathogenesis

Authors:

Caroline Grönwalla,b*, Khaled Amaraa, Uta Hardta, Akilan Krishnamurthya, Johanna Steena, Marianne Engströma, Meng Suna, A. Jimmy Ytterberga,c, Roman A. Zubarevc, Dagmar Scheel-Toellnerd, Jeffrey D. Greenbergb, Lars Klareskoga, Anca I. Catrina a, Vivianne Malmströma and Gregg J. Silvermanb

a) Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden

b) Division of Rheumatology, Department of Medicine, NYU School of Medicine, New York, NY, USA

c) Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden

d) Rheumatology Research Group, Centre for Translational Inflammation Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom

Oxidation-associated malondialdehyde (MDA) modification of proteins can generate immunogenic neo-epitopes that are recognized by autoantibodies. In health, IgM antibodies to MDA-adducts are part of the natural antibody pool, while elevated levels of IgG anti-MDA antibodies are associated with inflammatory and autoimmune conditions. Yet, in human autoimmune disease IgG anti-MDA responses have not been well characterized and their potential contribution to disease pathogenesis is not known. Here, we investigate MDA-modifications and anti-MDA-modified protein autoreactivity in rheumatoid arthritis (RA). While RA is primarily associated with autoreactivity to citrullinated antigens, we also observed increases in serum IgG anti-MDA in RA patients compared to controls. IgG anti-MDA levels significantly correlated with disease activity by DAS28-ESR and serum TNF-alpha, IL-6, and CRP. Mass spectrometry analysis of RA synovial tissue identified MDA-modified proteins and revealed shared peptides between MDA-modified and citrullinated actin and vimentin. Furthermore, relatively high frequency of anti-MDA autoreactivity among synovial B cells (3.5 % of memory B cells and 2.3% of plasma cells) was discovered when investigating recombinant monoclonal antibodies (mAbs) cloned from single B cells. Several clones were highly specific for MDA-modification with no cross-reactivity to other antigen modifications such as citrullination, carbamylation or 4-HNE-carbonylation. The mAbs recognized MDA-adducts in a variety of proteins including albumin, histone 2B, fibrinogen and vimentin. Interestingly, the most reactive clone, originated from an IgG1-bearing memory B cell, was encoded by near germline variable genes, and showed similarity to previously reported natural IgM. Other anti-MDA clones display somatic hypermutations and lower reactivity. Importantly, these anti-MDA antibodies had significant in vitro functional properties and induced enhanced osteoclastogenesis, while the natural antibody related high-reactivity clone did not. We postulate that these may represent distinctly different facets of anti-MDA autoreactive responses.

24. The prognostic value of IgA autoantibodies (rheumatoid factor and ACPA) for prediction of therapeutic responses to anti-TNF therapy in patients with rheumatoid arthritis

Daniela Sieghart1, Farideh Alasti1, Paul Studenic1, Thomas Horn2, Thomas Perkmann3, Daniel Aletaha1, Josef Smolen1, Günter Steiner1 1Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria; 2Phadia Austria GmbH, Thermo Fisher Scientific, Vienna, Austria; 3Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.

Background: Anti-citrullinated protein antibodies (ACPA) and rheumatoid factor (RF) are the most important serologic markers in rheumatoid arthritis (RA). These antibodies are predominantly of the IgM (RF) or IgG (ACPA) isotype while the added diagnostic and prognostic value of IgA autoantibodies has not been fully elucidated yet.

Objectives: To determine the prevalence of IgA-RF and IgA-ACPA in patients with RA and to investigate their predictive value regarding response to treatment with methotrexate (MTX) and TNF inhibitors.

Methods: A total of 255 patients were tested for the presence of IgA-RF and IgA-ACPA by EliA (Thermo Fisher Scientific). IgM-RF and IgG-ACPA were routinely measured by nephelometry and the anti-CCP EliA (Thermo Fisher Scientific), respectively. Therapeutic responses (as defined by SDAI50 and ACR20) to MTX and TNF blocking biologicals were analyzed in an inception cohort (n=104) who had started their DMARD therapy at our clinic.

Results: Among the 255 patients 114 (44.7%) were found to be IgA-RF positive: 10.5% of them were negative for IgM-RF, and 5.2% of patients were double negative for both IgM-RF and IgG-ACPA. IgA-ACPA were detected in 79 (31%) patients and apart from two patients all of them were also positive for IgG-ACPA. Together, 49% of the patients had at least one type of IgA antibody. Remarkably, the percentage of patients showing a SDAI50 response to TNF inhibitors was significantly lower in patients positive for IgA-RF and/or IgA-ACPA compared to IgM-RF and/or IgG-ACPA positive patients without IgA antibodies (58% vs 25% responders, p<0.0001). Similar results were obtained when ACR20 was used as response criteria. In contrast, the presence of IgA antibodies was not associated with the therapeutic response to methotrexate.

Conclusion: IgA-RF appears to have some added diagnostic value whereas IgA-ACPA only slightly increased the number of ACPA positive patients. However, both IgA-RF and IgA-ACPA appear to have considerable predictive value regarding the therapeutic response to TNF inhibitory biological drugs and could therefore help in further stratification of RA patients and therapeutic decision making.

Figure (optional): Percentages of SDAI50 responses to TNF inhibitors in patients with IgA-RF and/or IgA-ACPA compared to IgA negative patients (positive for IgM-RF and/or IgG-ACPA) and seronegative patients (104 patients in total). Among the 42 IgA positive patients, 20 were double positive, 13 solely positive for IgA-RF and 9 solely positive for IgA-ACPA.

25. Structural studies of citrullinated T-cell epitopes in rheumatoid arthritis

Anatoly Dubnovitsky1, Genadiy Kozhukh1, Tatiana Sandalova2, Adnane Achour2, Lars Klareskog1,

Vivianne Malmström1

1 Rheumatology Unit, Department of Medicine, Karolinska University Hospital in Solna, Karolinska Institutet, Stockholm, Sweden

2 Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Department of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden

Citrullinated proteins can be found in humans in the setting of both health and disease. Immune

tolerance to citrullinated proteins is lost in ACPA-positive RA patients. The strong genetic association

of ACPA-positive RA with certain HLA-DR alleles suggests an important role of MHC II-mediated

antigen presentation to CD4+ T cells in RA pathology. The conversion of arginine to citrulline leads to

a significant change in physicochemical properties of the amino acid side chain by removal of a

positive charge present on arginine residue. This can affect both interaction of the modified peptides

with HLA-DR as well as interaction of peptide-MHC complex with T-cell receptors. In the latter case, a

neo-epitope is created by citrullination. In order to provide better understanding of how citrullination

alters the above-mentioned interactions we have determined several crystal structures of peptide-

MHC complexes with arginine/citrulline residues located at different positions in the MHC peptide

binding cleft. The peptides we used in this study originate from the candidate autoantigens alpha-

enolase (amino acids 26-40 or 326-340) and cartilage intermediate layer protein (amino acids 297-

311).

Our crystal structures demonstrate that the arginine/citrulline residues, which were found in positions

P-1, P2 or P3 are all solvent exposed and can therefore interact with TCR. Moreover, the citrullination

did not alter the binding register of the peptide in the MHC cleft compared to arginine version of the

same peptide. Our data thus demonstrate that citrulline can occupy different positions in the MHC

binding cleft and how citrullination in these positions creates neo-antigens that can select for new

repertoire of T-cell receptors. Hereby the T cell reactivity to citrullinated peptides in RA is broader

than previously recognised.

26. Ex vivo analysis of autoantigen-specific T cell responses using a multi HLA-class II tetramer approach

Christina Gerstner1, Hannes Uchtenhagen1,2, Jennifer Pieper1, Anatoly Dubnovitsky3,4, Karolina Tandré5, Lars Rönnblom5, William Kwok2, Eddie A James6, Vivianne Malmström1

1 Rheumatology Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden

2 Translational Research Program, BRI at Virginia Mason, Seattle (WA), USA

3 Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden

4 Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden

5 Department of Medical Sciences, Rheumatology, Science for Life Laboratory, Uppsala, Sweden

6 Tetramer Core Facility, BRI at Virginia Mason, Seattle (WA), USA

HLA class II tetramers allow direct ex vivo enumeration and phenotypic characterization of antigen-specific T cells and have proven useful in different settings, e.g. allergy and vaccination.

Based on flow cytometry we developed a multi HLA-class II tetramer approach that renders it possible to look at numerous specificities simultaneously and is sufficiently sensitive at the same time as auto-reactive T cells are likely to be rare. We focused on validated citrullinated T cell epitopes previously studied in RA from �-enolase, fibrinogen and CILP.

To test robustness and sensitivity of our multi-tetramer assay, we performed repeated experiments on PBMCs of HLA-DRB1*04:01-positive healthy controls (HCs) as well as RA patients.

First, we examined the sensitivity of the panel by assessing PBMC from HCs where we could detect low frequencies of auto-reactive T cells (1-10 per million CD4), mostly displaying a naïve phenotype. This was in sharp contrast to the numbers of influenza-specific T cells in the same donors that were 10-20 fold higher in numbers and of memory phenotype. Next, we tested the robustness of the panel by running technical repeats of all HC samples, which yielded similar frequencies. Thereafter, we focused on RA patient PBMC obtained from repeated blood draws (2-4 weeks apart). Also in these samples, frequencies ranged between 1-10 tetramer-positive cells per million CD4+ T cells. Moreover, not all T cell specificities were present in all patients. Still, frequencies were found to be stable in the repeated blood draws in approximately half of the individuals, implicating that frequencies close to 1 cell per million CD4+ T cells is borderline of what we can stably detect. Importantly, the patient samples utilized were not taken from time points of active disease, where we hypothesize frequencies to be elevated.

In conclusion, we developed a sensitive tetramer panel allowing simultaneous enumeration and phenotypic characterization of antigen-specific T cells in ex vivo samples from RA patients that can be used for monitoring and in-depth studying of these cells during the course of disease and treatment in individual patients.

27. Comprehensive Spatial Transcriptomics analysis of inflammatory cell infiltrates in RA and SpA synovial tissue biopsies

Konstantin Carlberg1,2, Marina Korotkova2, Anca I. Catrina2, Joakim Lundeberg1, Vivianne Malmström2

1Science for Life Laboratory, Division of Gene Technology, KTH Royal Institute of Technology

2Karolinska Institute, Department of Medicine, Rheumatology Unit, Karolinska University Hospital

Background:

Rheumatoid Arthritis (RA) and Spondyloarthropathy (SpA) are two common chronic inflammatory joint diseases where recruitment of macrophages and T cells are commonly observed in the affected synovia. Current treatment regimes for the two disorders points to both common and unique features of disease, e.g. TNF blockade being widely used for both, while IL-17 targeting can be beneficial in SpA but less so for RA. Moreover, the genetic predisposition differs between RA and SpA with certain HLA-DR class II alleles associating to RA while SpA is genetically associated to the class I allele HLA-B27 and ER aminopeptidases.

In this study we profile the whole transcriptome of synovial tissue from RA and SpA samples while at the same time capture the spatial context of the gene expression patterns on each tissue section [1]. With this spatial transcriptomic (ST) method we retrieve higher granularity and can query the differences and commonalities of gene expression in the inflammatory infiltrates of affected joint tissue in RA and SpA.

Materials and methods:

Snap frozen synovial biopsies from three ACPA+ RA patients and three SpA patients undergoing joint replacement surgery was included. Sections of 7 μm representing a single layer of cells were placed on a barcoded ST slide and prepared as previously described [1]. Library preparation, sequencing and data processing were performed according to standard ST methodology [1][2][3].

Results:

Extracted RNA from the synovial biopsies had RIN values between 6.4 and 9.2. The complexity of the samples was mirrored by the numbers of detected genes which were 16-20,000 per tissue section. The number of barcoded spots covered by tissue was 300-500 while the number of unique genes per spot varied from 300 to 2,300. Hierarchical clustering based on data from the entire sections and the top 100 differentially expressed genes showed distinctive patterns for the two diseases. The platform allowed us to next re-analyze the data focusing only on the barcoded spots covered by inflammatory cell infiltrates and hereby zoom into the local cellular crosstalk e.g. between B and T cells.

Conclusion:

By using the spatial transcriptomic method, we have generated and analyzed unprecedented transcriptomic data on immunological tissue and compared two diseases revealing gene expression patterns that could not be inherited from homogenized RNA-seq samples.

28. Macrophages drive chronic active arthritis without involvement of T cells

Cecilia Hagert1,2, Outi Sareila1, Tiina Kelkka1,3, Kutty Selva Nandakumar4,5, Sirpa Jalkanen1, Rikard Holmdahl1,5

1.Medicity, University of Turku, Turku, Finland 2. The National Doctoral Programme in Informational and Structural Biology (ISB), Turku, Finland 3. The Turku Doctoral Programme of Biomedical Sciences (TuBS), Turku, Finland 4. School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China, 5. Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.

Abstract To analyze and evaluate chronic arthritis independently from the adaptive immunity we developed a novel, chronic, innate driven collagen antibody induced arthritis (CAIA) model. A four-monoclonal cocktail of anti-collagen type II antibodies initiate an active chronic arthritis, facilitated by mannan (Saccharomyces cerevisiae) injected on day five and sixty. Our model corresponds to the human rheumatoid arthritis both in its relapsing nature of the chronic phase and the inflammation caused by the infiltrating cells. This relapsing disease is driven by the complement pathway and ROS deficient macrophages. It is a model independent of FcγRIII, B- and T-cells, but a role for the innate immune system as driver of the chronic autoimmunity in this model is being suggested. In fact, our data illustrates an essential role for macrophage produced ROS to protect against the disease. On the other hand, the complement cascade seems to be promoting the disease, initiated via classical and/or alternate pathway but not through the lectin pathway. Our results, pinpoint macrophages and the

complement cascade as important drivers of active chronic arthritis, which is in contrast to the current dogma that arthritis is primarily driven by autoantibodies and T cells. Instead, this novel model specifies a role for the adaptive immunity in the beginning of arthritis, but at later stages of disease it will have less importance in driving the chronicity of the disease. This could suggest a need for an alternate approach while exploring new drug targets to attenuate arthritis.

29. THE MUTATED RNA SPLICING PROTEIN HNRNP-A3 IS A NOVEL AUTOANTIGEN IN SYSTEMIC RHEUMATIC DISEASES A LINK TO WARBURG EFFECT IN RA

B. Marklein1, M. Hansson2, G. Steiner3, G. Burmester1, K. Skriner1,*

1Department of Rheumatology and Clinical Immunology, CHARITÉ UNIVERSITY MEDICINE, Berlin, Germany, 26 Rheumatology Unit, Department of Medicine, Karolinska Institutet, Stockhom, Sweden, 32Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria

Background: The present study was undertaken to investigate novel anti-hnRNPs autoantibodies in rheumatic diseases.

Objectives: Novel mutated hnRNP A3 was cloned out of RA synovial tissue linking it directly to Warburg effect and lactate production in RA. Increased lactate production in RA synovia and tumors is klinked to alternative splicing process from PKM1 to PKM2 hnRNPs dependent

Methods: After immunobloting and 2D-gel-eletrophoresis of a semipurified hnRNP fraction two protein spots were sequenced and identified to be highly similar to hnRNPA3. The hnRNP A3 variants were cloned from RA synovial tissue, which identified the isoforms found on protein level. 3700 RA sera were screened for the presence of mutated anti-hnRNP A3 autoantibodies using recombinant proteins and peptides thereof. Binding of RNA to hnRNP A3 (MA3) and mutated citrullinated A3 peptides (MCA3) the epitope recognition was investigated. Expression of hnRNP A3 in synovial tissue was analysed by

Results: Autoantibodies to MA3 protein were detected in 13 % of RA (n=215) patients, in 9% SLE(n=154), in 27 % of MCTD patients (n=44/10) and in less than 5% of 129 patients with other rheumatic disorders but not at all in healthy controls on immunoblot. When using renaturated MA3 on ELISA 22% of RA patients were detected and 87 % of these patients had erosive arthritis. Same modification as in cancer cells were identified in synovial tissue and verified by MS and DNA sequencing. Using 2-3 citrullinated MCA3 peptides up to 81% of patients (n=150) with established and 67% (n=2926) of patients with an early RA with a specificity of 97% were detected. In early RA 27% and 25% in established RA of CCP2 negative and 93% of CCP2 positive patients were identified. By combining with the already established CCP2 and the new MCA3, 72% of early patients are positive. MCA3 autoantibodies predominantly occur (p <0.001) in an erosive, severe course of disease and approximately 60% of these patients receive a TNF alpha blocker within a year. MRL Lpr/lpr sera were hnRNP-A3 reactive and the antibody generation is Toll 7 and 9 dependent. Anti-hnRNP-A3-antibodies are directed to conformational RNA binding epitopes. Expression of hnRNP-A3 revealed the antigen to be overexpressed in RA synovial tissue.

Conclusions: Mutated hnRNP-A3 is as a novel Toll7/9 dependent autoantigen in systemic rheumatic diseases. These mutated proteins are major components of RNA and DNA containing alternative splicing complexes leading to the Warburg effect and and autoantibodies predominantly occurre in an erosive, and severe courses of RA.

30. NEW AUTOANTIGEN (JKTBP) PART OF STRESS GRANULES CLOSES THE SENSITIVITY GAP IN RHEUMATOID ARTHRITIS

B. Marklein1, M. Jenning1, K. Muenzer1, G. Burmester1, K. Skriner1,*

1Department of Rheumatology and Clinical Immunology, CHARITÉ UNIVERSITY MEDICINE, Berlin, Germany

Background: Rheumatoid arthritis triggers the formation of prion-like stress granules. To investigate which members of the heterogeneous nuclear ribonucleoprotein (hnRNP)-family, components of functionally important subcellular particles are targeted by autoantibodies from RA and other systemic rheumatic diseases.

Methods: Using a protein macroarray we identified JKTBP in humans and animal models of inflammatory rheumatic diseases. Bacterially expressed recombinant JKDBP proteins were used to confirm the obtained data. Epitope, TLR7/9 and MyD88 dependency was determined by ELISA. JKTBP expression in cultivated cells and synovial tissue was analysed by indirect immunofluorescence, immunoblot and immunohistochemistry.

Results: Anti-JKTBP autoantibodies were detected in 46% of the patients with systemic lupus erythematosus (n=103), in 20-30% of the patients with rheumatoid arthritis (n=286), in 10% of the patients with mixed connective tissue disease (n=20) or spondyloathropathy (n=20) , and in <10% of patients with other autoimmune disorders (n=382). Sera positive to JKTBP as well as hnRNP-B1, revealed nearly two thirds of the RF IgM/ CCP2-seronegative patients as early RA patients. Combining sensitivities to all autoantigens tested (JKTBP, AUF1, hnRNP-B1), it was possible to identify 92% of the early RA patients (n=91). In the MRL/lpr mouse model of SLE, mice deficient of MyD88 or TLR7/9 lacked anti-JKTBP autoantibodies, whereas mice deficient of SIGIRR/TIR8 showed enhanced anti-JKTBP autoantibody production. These results show that autoantibody generation against JKTBP, AUF1, hnRNP-B1 is dependent on TLR 7 and TLR9 like rheumatoid factor different to TLR 7 dependent generation of snRNPs. For all tested autoantigens either their titter or generation are dependent on the activation of innate immunity genes MyD88 and SIGIRR/TIR8 gene.

In localization, experiments anti-JKTBP autoantibodies specifically stained stress granules (SG) in the cytoplasm. Immunohistochemical studies revealed JKTBP to be highly expressed in SG in the cytoplasm of RA synovial tissue different from OA and normal control tissue

Conclusions: These data identify SG as targeted particle in RA and JKTBP as a novel autoantigen in RA, SLE patients and mouse models of inflammatory rheumatic diseases. In combination with the hnRNPs AUF1 and hnRNP-B1, JKTBP autoantibodies close the sensitivity gap in RA left by rheumatoid factor and anti-CCP2 antibodie

31. Impact of Toll-like receptor 9 in inflammatory arthritis and osteoclastogenesis

Anita Fischer*, Christina Böhm*, Marije Koenders†, Wim van den Berg†, Tobias Rothe‡, Gerhard Krönke‡, Diana Dudziak# and Günter Steiner*.

*Department of Rheumatology, Medical University of Vienna, Austria. †Department of Rheumatology, Radboud University Nijmegen Medical Centre, Netherlands. ‡Internal Medicine III and #Department of Dermatology, University of Erlangen-Nuremberg, Germany.

Background and Objectives. Release and insufficient removal of endogenous nucleic acids may be involved in triggering autoimmune reactions important in the initiation of systemic autoimmune diseases including rheumatoid arthritis (RA). Nucleic acid sensing molecules, such as the endosomal Toll-like receptors (TLRs) 7 and 9, have been linked to pathogenic autoimmune processes, but their role in RA is less clear. To gain more insight into the role of TLR9 in autoimmune arthritis, TLR9 inhibition was investigated in rats with pristane-induced arthritis (PIA). To further investigate TLR9 involvement, serum transfer arthritis was induced in TLR9 knock-out mice. Materials and Methods. Arthritis was induced in mice with arthritogenic K/BxN serum and in rats with the mineral oil pristane. Rats were treated with a TLR9 antagonist, starting before disease

induction. Arthritis was scored using established scoring systems, inflammation and bone erosion were quantified by histological analysis. The role of TLR9 in osteoclast differentiation and activation was investigated in vitro. Results. In the serum transfer model, which is independent of the adaptive immune system, arthritis severity was not changed in mice lacking a functional tlr9 gene. In PIA, which is T cell-dependent, the TLR9 antagonist reduced arthritis severity by ~50%. TLR9 inhibition led to reduced inflammation, bone erosion and cartilage degradation. However, when treatment was started after the onset of arthritis TLR9 inhibition did not influence disease severity. Treatment with the TLR9 antagonist influenced downstream signaling, mainly via NFκBp65, in lymph nodes. Remarkably, mRNA levels of TLR7 and TLR9 strongly differed in the course of in vitro osteoclastogenesis. Whereas TLR7 expression did not change throughout osteoclastogenesis, expression of TLR9 was higher in precursor cells than in mature osteoclasts and stimulation with a TLR9 agonist (CpG) completely inhibited osteoclastogenesis. Conclusions. Taken together, the results suggest a role for TLR9 in the T cell-dependent initiation phase of PIA and thus an important involvement of the DNA (CpG) recognizing TLR9 in the initiation of autoimmune arthritis and during osteoclastogenesis. The precise role(s) of TLR9 in the different stages of arthritis needs to be further elucidated in future experiments.

32. Expression and clinical significance of glutamate in the serum of patients with rheumatoid arthritis

LIANG Li-Jun1; DENG Zhao-Da2; ZAHO Qin1; SHEN Hai-Li3

Objective: To analyze the expression levels of glutamate(Glu), IL-17 and TNF-α in serum of

patients with rheumatoid arthritis(RA) and investigate the clinical significance of Glu in RA.

Method: A total of 34 RA patients were enrolled and 20 healthy subjects were enrolled as healthy

control group. Glu,IL-17and TNF-a levels were determined by enzyme-linked immunosorbent

assay(ELISA), serum Glu levels

were detected in 20 cases healthy controls by the same way. Pearson correlation analysis were used to

define the correlation of Glu,IL-17andTNF-α.RA patients were divided into bone erosion group and

non-bone erosion group and compare the differences of Glu,IL-17 and TNF-α in the two groups.

Results: The levels of Glu were significantly higher in RA group than healthy control

group[(14.16±7.76)μmol/L vs(11.31±1.08)μmol/L,P=0.003],and were positively correlated to IL-

17,TNF-a,visual analogue scale(VAS) scores,disease

activity score in 28 joints(DAS28) and tender joint count,however no correlation with ESR,CRP ,anti-

CCP antibody and swollen joint count. The levels of Glu, IL-17,TNF-a in serum were higher in the

bone-erosion group than the no bone-erosion group[(16.53±10.35)μmol/L,(273.44±114.31)pg/mL,

(169.95±92.58)pg/mL vs (14.79±4.90)μmol/L,(203.62±62.18)pg/mL,(107.49±56.42)pg/mL].

Conclusion: The levels of glutamate may be involved in the occurrence of RA inflammatory

pain,and has potential significance in predicting RA bone erosion.

1.Second School of Clinical Medicine, Lanzhou University,China;

2.Department of clinical medicine, Lanzhou University,China;

3.Department of Rheumatoid Immunology, Second Hospital of Lanzhou University, China

33. Stromal inflammatory memory mediates chronification of arthritis Christiane Reinwald1, Adela Rauschenberg1, Benjamin Wirth1, Vladimir Trajkovic2, Rabinder Prinhja3, Christopher D Buckley4, Andrew Filer4, Georg Schett1, and Markus H Hoffmann1

1Friedrich-Alexander University Erlangen-Nürnberg, Department of Internal Medicine 3, Universitätsklinikum Erlangen, Germany; 2Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, Serbia; 3Epigenetics DPU, Immuno-Inflammation Therapy Area Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, UK; 4Rheumatology Research Group, Institute of Inflammation and Ageing (IIA), University of Birmingham, UK.

Chronic inflammation causes severe collateral damage to the tissue that can be debilitating or life-threatening. The arthritic disorders Rheumatoid arthritis and gout are chronic, destructive inflammatory diseases often preceded by recurring, but spurious inflammatory attacks. In this project we aimed to characterize the mechanisms of the transition from acute inflammatory bouts to chronic inflammatory arthritis. Since local memory is likely mediated by stromal cells in the joint, we focused on the impact of synovial fibroblasts (SF) on chronification in arthritis in mice and humans. To model the early stages and the transition to the chronic phase in arthritis, we developed an animal model that relies on repeated injection of an inflammatory trigger into the paw, such as zymosan or monosodium urate crystals (MSU). In this model, a single exposure to zymosan/MSU led to acute but self-limiting arthritis in the respective paw. In contrast, arthritis exhibited a chronic course upon reinjection of the trigger into the same paw after resolution of the initial inflammation. Morphologically and functional comparison of SF isolated from paws that were exposed to the inflammatory trigger once or twice, respectively, revealed an elongated cell shape and higher interconnectivity in SF that had repeatedly been exposed (“trained” SF). This SF phenotype went along with increased invasiveness and higher cytokine production after exposure to the inflammatory trigger and seems to be epigenetically regulated. RNA-seq showed upregulation of the complement system and of integrins in trained SF. Blockade of integrins or complement abrogated interconnectivity and the development of inflammatory memory in vitro and in in vivo, respectively. We hypothesize that during the course of arthritis acute inflammation is gradually turned into a chronic state characterized by enhanced adhesion and higher interconnectivity between local SF. Another hallmark of chronic inflammation is the higher and prolonged reactivity upon re-exposure to an inflammatory trigger. We hypothesize that this effect is due to epigenetically regulated autoimmune memory formation, includes upregulation of integrins and the complement system, and is connected to the morphologic changes we observed and the interactions of different cells within an inflamed joint.

34.The short-term application of etanercept in the elderly in a follow-up trial in real world

WANG Xin1, XI Haipeng2, SU Jinhu3, LIANG Lijun1, WANG Xiaoyuan1, SHEN Haili1

Objective:Our aim was to compare therapeutic effects and safety of biological disease

modifying antirheumatic drugs (bDMARDs)amd conventional synthetic DMARD (csDMARD) in

elderly rheumatoid arthritis(RA) in a short-term follow-up investigation in Gansu area.

                                                            1 Department of Rheumatology,Lanzhou University Second Hospital,China 2 School of Information Science and Engineering, Lanzhou University,China 3 Department of Health Care ,Lanzhou Military Area Command of the Chinese People's Lliberation Army

Methods :100 patients were included and divided into 3 parts, elderly-onset RA(EORA, age at

disease onset>60)、non-elderly-onset RA(NEORA,age at disease onset<60,current age>60) and

younger-onset RA (YORA current age<60 ). 5 groups were named after the protocols,EORA-

bDMARDs(E-bD),NEORA-bDMARDs(NE-bD),EORA-csDMARDs(E-cD),NEORA-csDMARDs(NE-

cD),YORA-bDMARDs(Y-bD), each with 20 patients. Effectiveness and safety of the therapy were

compared among 5 groups.

Results:After 4 weeks following-up,there was a decrease in joint pain counts(E-bD9.30±7.09

,NE-bD7.15±6.68,Y-bD8.00±4.35, P<0.05),ESR(E-bD24.90±22.34,NE-bD26.15±22.33,Y-

bD29.00±17.88, P<0.05) and disease activity, in E-bD,NE-bD,Y-bD groups, especially in Y-bD(E-

bD1.87±1.34,E-cD1.42±0.76,NE-bD1.98±1.32,NE-cD1.06±0.81,Y-bD2.13±1.10, P>0.05),

however, the difference was not statistically significant. Data followed for 4 weeks showed a

larger drop of disease activity in E-bD and NE-bD groups than in E-cD and NE-cD groups,

P<0.05.After a median follow-up period of 8 weeks, the indicators of E-cD and NE-cD groups declined

compared with the baseline, but it was not statistically significant between these 2 groups

(P>0.05).The use of biological agents and csDMARDs carried a similar infection risk between EORA

and NEORA(P>0.05).It seemed Y-bD group had fewer adverse reactions than E-bD,NE-bD, but

differences were no significant(P>0.05).The adverse reactions had not statistical difference

between EORA and NEORA whatever the treatments was.

Conclusion :It would be a longer time for E-bD,NE-bD to get remissions than Y-bD groups,

meanwhile more untoward reaction might be found in E-bD,NE-bD groups. Biological agents had

a similar therapeutic effect on E-bD and NE-bD regardless of the age at disease onset and course.

Biologics has faster and better efficacy than csDMARDs in the same group, but there was

no statistics significant of adverse reactions bwtween them.The incidence of adverse events was not

associatedwith age at disease onset between E-cD and NE-cD groups.

35. Occupational Physical Workload and Development of Anti-Collagen Type II Antibodies in Rheumatoid Arthritis Patients

Pingling Zeng1 , Lars Alfredsson,1,2 Lars Klareskog,3 Mohammed Mullazehi4,

Saedis Saevarsdottir3, Camilla Bengtsson1, Johan Rönnelid4

1Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden

2Center for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden

3Rheumatology Unit, Department of Medicine at Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden

4Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden

Background/Purpose: We have previously observed an association between exposure to occupational physical workload (PW) and risk of developing rheumatoid arthritis (RA)[1].We posit that PW could impose mechanical stress on the joints leading to neo-epitope formation and immune activation. The major solid component of the articular cartilage is collagen type II (CII). Elevated level of anti-CII antibodies do not predate RA development, but are transiently found in 6-9% early RA patients. These antibodies are associated with acute RA onset and HLA-DRB1*01 and *03, yet predict good long-term prognosis[2]. We hypothesized that exposure to PW especially in the period closely predating RA diagnosis would associate with anti-CII positive RA but not anti-CII negative RA.

Methods: Data involving 1396 incident RA cases and 5935 controls from the Swedish population-based case-control study, Epidemiological Investigation of Rheumatoid Arthritis (EIRA), were analyzed. Information on self-reported exposure to occupational PW was collected through questionnaire. Anti-CII was measured using ELISA. The odds ratio (OR) with 95% confidence interval (CI) of developing anti-CII positive RA or anti-CII negative RA associated with PW exposure was calculated using logistic regression. Adjustment for sex, age, residential area, educational level, alcohol consumption, body mass index, cigarette smoking, silica exposure and occupational class did not substantially change the estimates.

Results: The ORs observed for the association between anti-CII positive RA and different types of PW ranged from 1.0 (95% CI, 0.4-2.4) to 2.3 (95% CI, 1.3-4.0). There was no difference in the ORs for PW exposure at diagnosis or five years earlier (table1). The ORs for the association between anti-CII negative RA and PW ranged from 1.0(95% CI, 0.8-1.3) to 1.8(95% CI, 1.5-2.1). No statistically significant difference was observed between the ORs for anti-CII positive RA and the ORs for anti-CII negative RA (all p-values > 0.2).Stratification for HLA-DRB1*01/*03 or symptom duration did not change the results.

Conclusion: Since physical workload is associated with rheumatoid arthritis irrespective of anti-CII status or the presence of anti-CII associated HLA alleles, and regardless of whether the timing of PW exposure was close to the appearance of anti-CII. We found no evidence suggesting an association between PW and development of anti-CII in RA.

Table 1 Odds ratios of developing anti-CII Positive RA and anti-CII negative RA among subjects exposed to occupational physical workload

Anti-CII Positive RA Anti-CII Negative RA

Baseline

Five Years Before Baseline

Baseline

Five Years Before Baseline

Types of Physical Workload

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

repetitive turning/

unexposed

24/1941

1.0(ref.)

26/2039

1.0(ref.)

318/1941

1.0(ref.)

326/2039

1.0(ref.)

bending exposed

38/2067

1.5 (0.9-2.6)

54/2531

1.7 (1.1-2.7)

513/2067

1.5 (1.3-1.8)

734/2531

1.8 (1.5-2.0)

repetitive hand/

unexposed

17/1351

1.0(ref.)

24/1546

1.0(ref.)

261/1351

1.0(ref.)

330/1546

1.0(ref.)

finger movements

exposed

45/2652

1.3 (0.7-2.3)

56/3020

1.2 (0.7-1.9)

571/2652

1.1 (1.0-1.3)

727/3020

1.1 (1.0-1.3)

carry >10kg

unexposed

33/2418

1.0(ref.)

38/2496

1.0(ref.)

420/2418

1.0(ref.)

434/2496

1.0(ref.)

exposed

29/1585

1.5 (0.9-2.5)

42/2077

1.5 (0.9-2.3)

412/1585

1.5 (1.3-1.7)

625/2077

1.7 (1.5-2.0)

precision work

unexposed

56/3584

1.0(ref.)

72/4061

1.0(ref.)

742/3584

1.0(ref.)

934/4061

1.0(ref.)

exposed

6/398

1.0 (0.4-2.4)

8/487

1.0 (0.5-2.1)

88/398

1.0 (0.8-1.3)

124/487

1.1 (0.9-1.3)

hands below

unexposed

54/3448

1.0(ref.)

66/3827

1.0(ref.)

666/3448

1.0(ref.)

800/3827

1.0(ref.)

knee level

exposed

8/546

1.0 (0.5-2.1)

14/736

1.2 (0.7-2.1)

165/546

1.5 (1.2-1.8)

259/736

1.6 (1.4-1.9)

vibration unexposed

52/3492

1.0(ref.)

68/3927

1.0(ref.)

692/3492

1.0(ref.)

862/3927

1.0(ref.)

exposed

10/499

1.7 (0.8-3.6)

12/634

1.4 (0.7-2.7)

139/499

1.3 (1.1-1.7)

197/634

1.4 (1.2-1.7)

hands above

unexposed

44/3331

1.0(ref.)

56/3675

1.0(ref.)

622/3331

1.0(ref.)

728/3675

1.0(ref.)

shoulder level

exposed

18/668

2.3 (1.3-4.0)

24/893

1.9 (1.2-3.2)

211/668

1.6 (1.4-2.0)

330/893

1.8 (1.5-2.1)

OR adjusted for age, sex and residential area. Baseline and five years before baseline refer to the occupational physical workload exposure status at the time of diagnosis and

at 5 years prior to diagnosis respectively. OR, odds ratio; anti-CII, anti-collagen type II anti-bodies ; RA , rheumatoid arthritis; 95% CI, 95% confidence interval

36. Association between Occupational Physical Workloads and Development of Anti-Collagen Type II Antibodies in Rheumatoid Arthritis Patients

Pingling Zeng1 , Lars Alfredsson,1,2 Lars Klareskog,3 Saedis Saevarsdottir3 ,Camilla Bengtsson1, Johan Rönnelid4

1Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden

2Center for Occupational and Environmental Medicine, Stockholm County Council, Stockholm, Sweden

3Rheumatology Unit, Department of Medicine at Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden

4Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala Sweden

Background/ Purpose: We have previously observed an association between exposure to occupational physical workloads (PW) and risk of developing rheumatoid arthritis (RA)[1].We posit that PW could impose excessive mechanical stress on the proteins of the joints which may lead to the formation of neo-epitopes that activate the immune system. Mechanical stress loaded on the joints is facilitated by the articular cartilage. The major solid component of the articular cartilage is collagen type II (CII). Elevated level of antibodies against CII is found in around 3-27% of RA patients[2]. Given this background, we hypothesized that the development of anti-CII antibodies in RA patients may be associated with exposure to physical workload at work.

Methods: Data involving 1396 incident RA cases and 5935 controls from the Swedish population case-control study, Epidemiological Investigation of Rheumatoid Arthritis (EIRA), were analyzed. Information on self-reported exposure to occupational PW was collected through questionnaire. The level of anti-CII antibody in the serum samples was measured. The odds ratio (OR) with 95% confidence interval (95% CI) of developing anti-CII positive RA or anti-CII negative RA associated with the exposure to PW was calculated using logistic regression. Adjustment for sex, age, residential area, educational level, alcohol consumption, body mass index, cigarette smoking, silica exposure, symptom duration and occupational class did not substantially change the estimates.

Results: The ORs observed for the association between anti-CII positive RA and different types of physical workload ranged from 1.0 (95%CI, 0.4-2.4) to 2.3 (95%CI, 1.3-4.0) (table 1). The ORs for the association between anti-CII negative RA and physical workloads ranged from 1.0(95%CI, 0.8-1.3) to 1.8(95%CI, 1.5-2.1). No statistically significant difference between the ORs for anti-CII positive RA and the ORs for anti-CII negative RA was observed (all p-values > 0.2).

Conclusion: Since physical workload is associated with rheumatoid arthritis regardless of whether anti-collagen type II antibodies are present, we found no evidence suggesting an association between occupational physical workload exposure and development of anti-collagen type II antibodies in rheumatoid arthritis patients.

Table 1 Odds ratios of developing anti-CII Positive RA and anti-CII negative RA among subjects exposed to occupational physical workload

Anti-CII Positive RA Anti-CII Negative RA

Baseline

Five Years Before Baseline

Baseline

Five Years Before Baseline

Types of Physical Workload

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

cases/ controls

OR (95%CI)

repetitive turning/

unexposed

24/1941

1.0(ref.)

26/2039

1.0(ref.)

318/1941

1.0(ref.)

326/2039

1.0(ref.)

bending exposed

38/2067

1.5 (0.9-2.6)

54/2531

1.7 (1.1-2.7)

513/2067

1.5 (1.3-1.8)

734/2531

1.8 (1.5-2.0)

repetitive hand/

unexposed

17/1351

1.0(ref.)

24/1546

1.0(ref.)

261/1351

1.0(ref.)

330/1546

1.0(ref.)

finger movements

exposed

45/2652

1.3 (0.7-2.3)

56/3020

1.2 (0.7-1.9)

571/2652

1.1 (1.0-1.3)

727/3020

1.1 (1.0-1.3)

carry >10kg

unexposed

33/2418

1.0(ref.)

38/2496

1.0(ref.)

420/2418

1.0(ref.)

434/2496

1.0(ref.)

exposed

29/1585

1.5 (0.9-2.5)

42/2077

1.5 (0.9-2.3)

412/1585

1.5 (1.3-1.7)

625/2077

1.7 (1.5-2.0)

precision work

unexposed

56/3584

1.0(ref.)

72/4061

1.0(ref.)

742/3584

1.0(ref.)

934/4061

1.0(ref.)

exposed

6/398

1.0 (0.4-2.4)

8/487 1.0

(0.5-2.1)

88/398

1.0 (0.8-1.3)

124/487

1.1 (0.9-1.3)

hands below

unexposed

54/3448

1.0(ref.)

66/3827

1.0(ref.)

666/3448

1.0(ref.)

800/3827

1.0(ref.)

knee level

exposed

8/546

1.0 (0.5-2.1)

14/736

1.2 (0.7-2.1)

165/546

1.5 (1.2-1.8)

259/736

1.6 (1.4-1.9)

vibration unexposed

52/3492

1.0(ref.)

68/3927

1.0(ref.)

692/3492

1.0(ref.)

862/3927

1.0(ref.)

exposed

10/499

1.7 (0.8-3.6)

12/634

1.4 (0.7-2.7)

139/499

1.3 (1.1-1.7)

197/634

1.4 (1.2-1.7)

hands above

unexposed

44/3331

1.0(ref.)

56/3675

1.0(ref.)

622/3331

1.0(ref.)

728/3675

1.0(ref.)

shoulder level

exposed

18/668

2.3 (1.3-4.0)

24/893

1.9 (1.2-3.2)

211/668

1.6 (1.4-2.0)

330/893

1.8 (1.5-2.1)

OR adjusted for age, sex and residential area. Baseline and five years before baseline refer to the occupational physical workload exposure status at the time of diagnosis and

at 5 years prior to diagnosis respectively. OR, odds ratio; anti-CII, anti-collagen type II ; RA , rheumatoid arthritis; 95% CI, 95% confidence interval

37. Is Leukotoxin A produced by Aggregatibacter Actynomycetemcomitans Important For Initiating Autoimmune Responses Underlying Rheumatoid Arthritis?

Mikhail Volkov1, Jacqueline Dekkers1, Bruno G. Loos2, Sergio Bizzarro2, Thomas W.J. Huizinga1, Helle A. Praetorius3, René E.M. Toes1, Diane van der Woude1

1. Department of Rheumatology, Leiden University Medical Center, Leiden, the Netherlands

2. Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, the Netherlands

3. Department of Biomedicine, Aarhus University, Aarhus, Denmark

Background: In a recent publication in Science Translational Medicine, Konig et al. (2016) describe a potential explanation for the link between periodontal infection and RA. They identify a specific periodontitis-associated bacterium: Aggregatibacter actinomycetemcomitans (Aa), which via its pore-forming toxin (leukotoxin A: LtxA) can dysregulate the activity of citrullinating enzymes in neutrophils. Furthermore, the authors report that the risk conferred by the most important genetic risk factor for RA: the HLA-DRB1 shared epitope (HLA SE) alleles, was limited to RA patients who had been exposed to Aa as determined by seropositivity to LtxA. Aiming to replicate their findings, we focused on two main questions: 1) is the increased exposure to Aa as measured by the presence of anti-LtxA-antibodies specific for RA, or also present in other forms of inflammatory arthritis? 2) can we replicate the finding that the association between HLA SE alleles and ACPA-positive RA is limited to the anti-LtxA-positive subset?

Methods: We established an ELISA against purified LtxA (acquired from the same source as in the original article) and tested sera from 594 patients from the Leiden Early Arthritis Clinic with various diagnoses, including RA, OA, SpA, PsA, sarcoidosis, and gout. Serial dilutions of a mix of 3 strongly positive RA patients were used as a standard, and the lowest point of the linear part of the standard curve (2000 AU/ml) was defined as the cut-off.

Results: Anti-LtxA antibodies could be found in a substantial proportion of RA patients, but also in patients with other forms of arthritis. Within RA patients, there was no association with the presence of HLA SE alleles and/or ACPA, in contrast to the previous findings.

Conclusion: Although microbial influences may well be important in the development of RA, our results do not support a key role of exposure to LtxA originating from the periodontal pathogen Aa in linking the effect of the HLA SE alleles and periodontal disease to anti-citrullinated protein autoimmunity in RA.

38. Free secretory component among patients with recent-onset RA and patients with ACPA-positive musculo-sceletal pain

Klara Martinsson, Michael Ziegelasch, Jan Cedergren, Per Eriksson, Vladimir Klimovich*, Åsa Reckner-Olsson, Marina Samoylovich*, Christopher Sjöwall, Thomas Skogh, Jonas Wetterö & Alf Kastbom.

Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden, *Russian Research Center for Radiology and Surgical Technologies, St. Petersburg, Russia

Background: Emerging evidence suggests that mucosal immunity is important for development of rheumatoid arthritis (RA). Secretory component (SC) is expressed on mucosal epithelial cells and involved in formation of secretory IgA (sIgA). During this process, SC may be shed, forming free SC. This project aimed to determine if free circulating SC is elevated in RA or during its development. We also addressed the prognostic value of circulating SC levels among patients with recent-onset RA.

Materials & Methods: Two inception cohorts were studied, i.e. TIRA2 (timely intervention in RA) comprising 498 recent-onset RA patients (symptom duration <12 months) with clinical and radiological follow-up during 3 years, and the TIRx cohort (Xtra timely RA follow-up) including 110 patients with circulating IgG anti-citrullinated peptide antibodies (IgG-ACPA) and musculo-skeletal pain, but ≤1 joint with palpable synovitis (the majority having none). Free SC was analysed in serum using a sandwich ELISA. Controls comprised of blood donors (n=100) and patients with ankylosing spondylitis (AS), ANCA-associated vasculitis (AAV) or systemic lupus erythematosus (SLE) (total n=169).

Results: Free SC was elevated in TIRA2 patients compared to all controls (p<0.01). The proportion of patients testing positive was higher among IgG-ACPA positive patients compared to IgG-ACPA negative patients (p<0.001). Free SC levels correlated with sIgA-ACPA in serum (r=0.55; p<0.001), IgG-ACPA (r=0.38; P<0.001) and smoking. 3 months after inclusion, the levels of free SC had declined slightly, and the change in SC level correlated with change in disease activity score (DAS28; r=0.12; p=0.006). Free SC levels were increased among TIRx patients compared to blood donors, AAV patients, and SLE patients, but not to AS patients. No significant difference in free SC levels was detected in those who had palpable synovitis at baseline, or developed RA within 1 year.

Conclusion: Free SC levels are higher among recent-onset RA patients compared to other inflammatory diseases and associate with presence and levels of ACPA. Furthermore, free SC was elevated among IgG-ACPA positive patients prior to arthritis development. These findings provide yet another link between ACPA production and mucosal surfaces early in RA development.

39. LILRA3 contributes to osteoclastogenesis independently or synergy with RANKL

Jianping Guo, Mengru Liu, Yan Du, Yuxuan Wang, Yundong Zou, Fanlei Hu, Zhanguo Li

Department of Rheumatology and Immunology, Peking University People's Hospital,

Beijing, China

Objective Leukocyte immunoglobulin-like receptor A3 (LILRA3) is a secreted protein belongs to

LILR family. Our research group previously reported that the functional LILRA3 is a novel genetic risk

for multiple autoimmune diseases including rheumatoid arthritis (RA). LILRA3 had an impact on joint

destruction, especially in early ACPA-positive RA. In present study, we aimed to investigate the effect

of the variant on osteoclast differentiation.

Methods Expression of LILRA3 in serum and synovial fluid (SF) from patients with RA, OA, and

healthy controls were measured by ELISA. RAW264.7 cells or PBMCs from RA, OA patients and

healthy controls were cultured or transfected with recombinant LILRA3 protein in presence or absence

of RANKL and M-CSF. Expression of osteoclast-related markers, including TRAP, CTSK, MMP-9 were

measured by q-PCR, IHC and western blotting method. Bone resorption activity of osteoclasts was

determined by bone slices stained with toluidine blue. Three MAPK family signaling members

including ERK, JNK and p38, and NF-B signaling pathway activities were assessed by western

blotting.

Results Compared to healthy controls, the levels of serum LILRA3 were increased in patients with RA

and OA (p<0. 01). LILRA3 was specifically expressed in SF from RA but not OA patients (p<0.001).

The serum LILRA3 level was positively correlated with Sharp-vander Heijde scores in RA patients

(r=0.36, p=0.003). LILRA3 promoted osteoclast differentiation independently or synergy with

RANKL in RAW264.7 cells or human PBMCs through ERK, JNK and NF-B, but not p38 signaling

pathways.

Conclusion Our data indicate that LILRA3 may serve a potent pro-inflammatory factor participated

in osteoclast differentiation through MAPK family and NF-B signaling pathways in RA development.

40. LOW RATES OF RADIOGRAPHIC PROGRESSION OF STRUCTURAL JOINT

DAMAGE OVER 2 YEARS OF BARICITINIB TREATMENT IN PATIENTS WITH

RHEUMATOID ARTHRITIS

Authors: D. van der Heijde1, M. Schiff2, Y. Tanaka3, R. Klar4, L. Xie5, G. Meszaros5, T. Ishii5, M.

Casillas5, R. Ortmann5, P. Emery6, E. Larsson7 (Non-Author Presenter)

Affiliations: 1Leiden University Medical Center, Leiden, Netherlands, 2University of Colorado,

Denver, United States, 3School of Medicine, Kitakyushu, Japan, 4Quintiles IMS Holdings, Inc,

Durham, 5Eli Lilly and Company, Indianapolis, United States, 6University of Leeds, Leeds, United

Kingdom,7Eli Lilly Sweden AB, Solna, Sweden

Background: In a ph3 study, baricitinib (bari) inhibited progression of radiographic joint damage for

up to 1 year in patients (pts) with active rheumatoid arthritis (RA) who were DMARD-naïve or who

had an inadequate response to conventional synthetic DMARDs (csDMARD-IR).

Objectives: To evaluate radiographic progression of structural joint damage in pts with RA over 2

years of treatment.

Methods: Upon completion of a bari ph 3 study, pts could enter a long-term extension (LTE) study, in

which they continued to receive the same bari dose as in the original ph3 study. At 52 wks, DMARD-

naïve pts receiving methotrexate (MTX) or combination therapy (bari 4mg + MTX) were switched to

bari 4mg monotherapy; MTX-IR pts receiving adalimumab (ADA) were switched to bari 4mg on

background MTX. At 24 wks, csDMARD-IR pts receiving placebo (PBO) were switched to 4mg on

background csDMARD. Radiographs at baseline, year 1 and year 2 were scored using the van der

Heijde modified total sharp score (mTSS). Data are least squares mean change from baseline using

mixed model repeated measures on observed data.

Results: Of all pts randomised, 82.5% entered the LTE, and 87.6% of those could be entered in this

analysis. At year 2, progression was significantly lower with initial bari (including monotherapy) vs.

initial MTX in DMARD-naïve pts. In MTX/csDMARD-IR pts, progression with initial bari was

significantly lower than initial PBO, and similar to initial ADA.

Conclusions: Treatment with once-daily oral bari resulted in low rates of radiographic progression

for up to 2 years. Pts starting with bari showed progression that was significantly less than those

starting with PBO or MTX, and comparable to those starting with ADA. The most robust benefit was

seen with the 4mg dose.

This abstract was previously presented at EULAR 2017, Madrid, 14th-17th June and published in the

Annals of the Rheumatic Diseases, June 2017, Volume 76 (Supplement 2), page 510-511.

Disclosure of Interest: D. van der Heijde Consultant for: AbbVie, Amgen, Astellas, AstraZeneca,

BMS, Boeringer Ingelheim, Celgene, Daiichi Sankyo, Eli Lilly and Company, Galapagos, Gilead,

Janssen, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi-Aventis, UCB, Employee of: Director of

Imaging Rheumatology bv, M. Schiff Consultant for: Abbvie, BMS, Eli Lilly and Company, Johnson &

Johnson, Speakers bureau: Abbvie, Y. Tanaka Grant/research support from: Mitsubishi-Tanabe,

Takeda, Daiichi-Sankyo, Chugai, BMS, MSD, Astellas, Abbvie, Eisai, Speakers bureau: Abbvie, Chugai,

Daiichi-Sankyo, BMS, Mitsubishi-Tanabe, Astellas, Takeda, Pfizer, Teijin, Asahi-kasei, YL Biologics,

Sanofi, Janssen, Eli Lilly and Company, GlaxoSmithKline, R. Klar Employee of: Quintiles IMS

Holdings, Inc., L. Xie Employee of: Eli Lilly and Company, G. Meszaros Employee of: Eli Lilly and

Company, T. Ishii Employee of: Eli Lilly and Company, M. Casillas Employee of: Eli Lilly and

Company, R. Ortmann Employee of: Eli Lilly and Company, P. Emery Consultant for: Pfizer, MSD,

Abbvie, BMS, UCB, Roche, Novartis, Samsung, Sandoz, Eli Lilly and Company.