Plasma interferon-alpha is associated with double-positivity for autoantibodies but is not a predictor of remission in early rheumatoid arthritis-a spin-off study of the NORD-STAR randomized clinical trial

Marit Stockfelt, Anna-Carin Lundell, Merete Lund Hetland, Mikkel Østergaard, Till Uhlig, Marte Schrumpf Heiberg, Espen A Haavardsholm, Michael T Nurmohamed, Jon Lampa, Dan Nordström, Kim Hørslev Petersen, Bjorn Gudbjornsson, Gerdur Gröndal, Jonathan Aldridge, Kerstin Andersson, Kaj Blennow, Henrik Zetterberg, Ronald van Vollenhoven, Anna Rudin, Marit Stockfelt, Anna-Carin Lundell, Merete Lund Hetland, Mikkel Østergaard, Till Uhlig, Marte Schrumpf Heiberg, Espen A Haavardsholm, Michael T Nurmohamed, Jon Lampa, Dan Nordström, Kim Hørslev Petersen, Bjorn Gudbjornsson, Gerdur Gröndal, Jonathan Aldridge, Kerstin Andersson, Kaj Blennow, Henrik Zetterberg, Ronald van Vollenhoven, Anna Rudin

Abstract

Background: The type I interferon (IFN) gene signature is present in a subgroup of patients with early rheumatoid arthritis (RA). Protein levels of IFNα have not been measured in RA and it is unknown whether they associate with clinical characteristics or treatment effect.

Methods: Patients with early untreated RA (n = 347) were randomized to methotrexate combined with prednisone, certolizumab-pegol, abatacept, or tocilizumab. Plasma IFNα protein levels were determined by single molecular array (Simoa) before and 24 weeks after treatment initiation and were related to demographic and clinical factors including clinical disease activity index, disease activity score in 28 joints, swollen and tender joint counts, and patient global assessment.

Results: IFNα protein positivity was found in 26% of the patients, and of these, 92% were double-positive for rheumatoid factor (RF) and anti-citrullinated protein antibodies (ACPA). IFNα protein levels were reduced 24 weeks after treatment initiation, and the absolute change was similar irrespective of treatment. IFNα protein positivity was associated neither with disease activity nor with achievement of CDAI remission 24 weeks after randomization.

Conclusion: IFNα protein positivity is present in a subgroup of patients with early RA and associates with double-positivity for autoantibodies but not with disease activity. Pre-treatment IFNα positivity did not predict remission in any of the treatment arms, suggesting that the IFNα system is distinct from the pathways of TNF, IL-6, and T-cell activation in early RA. A spin-off study of the NORD-STAR randomized clinical trial, NCT01491815 (ClinicalTrials), registered 12/08/2011, https://ichgcp.net/clinical-trials-registry/NCT01491815 .

Conflict of interest statement

MS, ACL, TU, MTN, JL, KHP, GG, JA, KA, and AR have no competing interests to declare. MLH has received research grants from Abbvie, Biogen, BMS, Celltrion, Eli-Lilly, Janssen Biologics B.V, Lundbeck Fonden, MSD, Pfizer, Roche, Samsung Bioepis, Sandoz, and Novartis; chairs the steering committee of the Danish Rheumatology Quality Registry (DANBIO), which receives public funding from the hospital owners and funding from pharmaceutical companies; and co-chairs EuroSpA, which generates real-world evidence of treatment of psoriatic arthritis and axial spondyloarthritis based on secondary data and is partly funded by Novartis. MØ has received research grants from Abbvie, BMS, Merck, Celgene, and Novartis, and speaker and/or consulting fees from Abbvie, BMS, Boehringer-Ingelheim, Celgene, Eli-Lilly, Hospira, Janssen, Merck, Novartis, Novo, Orion, Pfizer, Regeneron, Roche, Sandoz, Sanofi, and UCB. MSH has received speaker’s honoraria from Lilly and Roche over the last 4 years outside the submitted work. EAH has received grants from the Norwegian Regional Health Authorities and The South-Eastern Norway Regional Health Authority during the conduct of the NORD-STAR study, and speaker and/or consulting fees from Pfizer, AbbVie, Celgene, Novartis, Janssen, Gilead, Eli-Lilly, and UCB outside the submitted work. DN has received consulting fees from AbbVie, BMS, MSD, Novartis, Pfizer, Roche, and UCB. BG has received speaking fees from Amgen and Novartis. KB has served as a consultant, at advisory boards or at data monitoring committees for Abcam, Axon, Biogen and JOMDD/Shimadzu, Julius Clinical, Lilly, MagQu, Novartis, Roche Diagnostics, and Siemens Healthineers and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside the submitted work). HZ has served at scientific advisory boards for Eisai, Denali, Roche Diagnostics, Wave, Samumed, Siemens Healthineers, Pinteon Therapeutics, Nervgen, AZTherapies, and CogRx; has given lectures in symposia sponsored by Cellectricon, Fujirebio, Alzecure, and Biogen; and is a co-founder of Brain Biomarker Solutions in Gothenburg AB (BBS), which is a part of the GU Ventures Incubator Program (outside submitted work). RV has received research and educational support (grants) from BMS, GSK, Lilly, Pfizer, Roche, and UCB and consultancy and/or speaking fees from AbbVie, AstraZeneca, Biogen, Biotest, BMS, Galapagos, Gilead, GSK, Janssen, Pfizer, Sanofi, Servier, UCB, and Vielabio.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Elevated IFNα protein levels at baseline in early RA. IFNα protein levels in plasma from patients with early RA before treatment initiation in four treatment arms, methotrexate + prednisone, methotrexate + TNFi, methotrexate + CTLA-4Ig, and methotrexate + IL-6Ri. The dotted line denotes the cut-off for IFNα positivity (136 fg/mL). MTX (methotrexate), TNFi (certolizumab-pegol), CTLA-4Ig (abatacept), and IL-6Ri (tocilizumab). Kruskal-Wallis test followed by Dunn’s multiple comparison test
Fig. 2
Fig. 2
IFNα protein levels are reduced after treatment initiation with conventional and biologic treatment strategies. IFNα protein levels in plasma from patients with early RA before (d1) and 24 weeks after treatment initiation (w24) with A methotrexate + prednisone (n = 85), B methotrexate + TNFi (n = 87), C methotrexate + CTLA-4Ig (n = 91), and D methotrexate + IL-6Ri (n = 82). Wilcoxon matched-pairs signed rank test. E Absolute difference in IFNα plasma protein levels between week 24 and day 1 in four treatment arms. MTX (methotrexate), TNFi (certolizumab-pegol), CTLA-4Ig (abatacept), and IL-6Ri (tocilizumab). Kruskal-Wallis test followed by Dunn’s multiple comparison test
Fig. 3
Fig. 3
Baseline IFNα protein levels do not predict remission after treatment. Baseline IFNα protein levels in plasma from patients with early RA, stratified according to CDAI 24 weeks after treatment initiation; in remission (CDAI 0–2.8), low disease activity (CDAI 2.9–10.0), and moderate/high disease activity (CDAI 10.1–76.0) with A all treatments, B methotrexate + prednisone, C methotrexate + TNFi, D methotrexate + CTLA-4Ig, and E methotrexate + IL-6Ri. MTX (methotrexate), TNFi (certolizumab-pegol), CTLA-4Ig (abatacept), IL-6Ri (tocilizumab). Kruskal-Wallis test followed by Dunn’s multiple comparison test

References

    1. Sugiyama D, Nishimura K, Tamaki K, Tsuji G, Nakazawa T, Morinobu A, Kumagai S. Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis. 2010;69(1):70–81. doi: 10.1136/ard.2008.096487.
    1. Han SW, Lee WK, Kwon KT, Lee BK, Nam EJ, Kim GW. Association of polymorphisms in interferon regulatory factor 5 gene with rheumatoid arthritis: a metaanalysis. J Rheumatol. 2009;36(4):693–697. doi: 10.3899/jrheum.081054.
    1. Remmers EF, Plenge RM, Lee AT, Graham RR, Hom G, Behrens TW, de Bakker PIW, le JM, Lee HS, Batliwalla F, Li W, Masters SL, Booty MG, Carulli JP, Padyukov L, Alfredsson L, Klareskog L, Chen WV, Amos CI, Criswell LA, Seldin MF, Kastner DL, Gregersen PK. STAT4 and the risk of rheumatoid arthritis and systemic lupus erythematosus. N Engl J Med. 2007;357(10):977–986. doi: 10.1056/NEJMoa073003.
    1. Castañeda-Delgado JE, Bastián-Hernandez Y, Macias-Segura N, Santiago-Algarra D, Castillo-Ortiz JD, Alemán-Navarro AL, et al. Type I interferon gene response is increased in early and established rheumatoid arthritis and correlates with autoantibody production. Front Immunol. 2017;8:285. doi: 10.3389/fimmu.2017.00285.
    1. Rodríguez-Carrio J, Alperi-López M, López P, Ballina-García FJ, Suárez A. Heterogeneity of the type I interferon signature in rheumatoid arthritis: a potential limitation for its use as a clinical biomarker. Front Immunol. 2018;8:2007. doi: 10.3389/fimmu.2017.02007.
    1. Cantaert T, van Baarsen LG, Wijbrandts CA, Thurlings RM, van de Sande MG, Bos C, van der Pouw TK, Verweij CL, Tak PP, Baeten DL. Type I interferons have no major influence on humoral autoimmunity in rheumatoid arthritis. Rheumatology. 2010;49(1):156–166. doi: 10.1093/rheumatology/kep345.
    1. de Jong TD, Blits M, de Ridder S, Vosslamber S, Wolbink G, Nurmohamed MT, Verweij CL. Type I interferon response gene expression in established rheumatoid arthritis is not associated with clinical parameters. Arthritis Res Ther. 2016;18(1):290. doi: 10.1186/s13075-016-1191-y.
    1. McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nature Reviews Immunology. 2015;15(2):87–103. doi: 10.1038/nri3787.
    1. Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA, Jallal B, Yao Y. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis. 2011;70(11):2029–2036. doi: 10.1136/ard.2011.150326.
    1. Lübbers J, Brink M, van de Stadt LA, Vosslamber S, Wesseling JG, van Schaardenburg D, Rantapää-Dahlqvist S, Verweij CL. The type I IFN signature as a biomarker of preclinical rheumatoid arthritis. Ann Rheum Dis. 2013;72(5):776–780. doi: 10.1136/annrheumdis-2012-202753.
    1. Cooles FAH, Anderson AE, Lendrem DW, Norris J, Pratt AG, Hilkens CMU, et al. The interferon gene signature is increased in patients with early treatment-naive rheumatoid arthritis and predicts a poorer response to initial therapy. J Allergy Clin Immunol. 2018;141(1):445–8.e4. doi: 10.1016/j.jaci.2017.08.026.
    1. Sekiguchi N, Kawauchi S, Furuya T, Inaba N, Matsuda K, Ando S, Ogasawara M, Aburatani H, Kameda H, Amano K, Abe T, Ito S, Takeuchi T. Messenger ribonucleic acid expression profile in peripheral blood cells from RA patients following treatment with an anti-TNF-alpha monoclonal antibody, infliximab. Rheumatology. 2008;47(6):780–788. doi: 10.1093/rheumatology/ken083.
    1. van Baarsen LG, Wijbrandts CA, Rustenburg F, Cantaert T, van der Pouw Kraan TC, Baeten DL, et al. Regulation of IFN response gene activity during infliximab treatment in rheumatoid arthritis is associated with clinical response to treatment. Arthritis Res Ther. 2010;12(1):R11. doi: 10.1186/ar2912.
    1. Wright HL, Thomas HB, Moots RJ, Edwards SW. Interferon gene expression signature in rheumatoid arthritis neutrophils correlates with a good response to TNFi therapy. Rheumatology. 2015;54(1):188–193. doi: 10.1093/rheumatology/keu299.
    1. Sanayama Y, Ikeda K, Saito Y, Kagami S-I, Yamagata M, Furuta S, et al. Prediction of therapeutic responses to tocilizumab in patients with rheumatoid arthritis: biomarkers identified by analysis of gene expression in peripheral blood mononuclear cells using genome-wide DNA microarray. Arthritis & Rheumatology. 2014;66(6):1421–1431. doi: 10.1002/art.38400.
    1. de Jong TD, Sellam J, Agca R, Vosslamber S, Witte BI, Tsang ASM, et al. A multi-parameter response prediction model for rituximab in rheumatoid arthritis. Joint Bone Spine. 2018;85(2):219–226. doi: 10.1016/j.jbspin.2017.02.015.
    1. Raterman HG, Vosslamber S, de Ridder S, Nurmohamed MT, Lems WF, Boers M, van de Wiel M, Dijkmans BAC, Verweij CL, Voskuyl AE. The interferon type I signature towards prediction of non-response to rituximab in rheumatoid arthritis patients. Arthritis Res Ther. 2012;14(2):R95. doi: 10.1186/ar3819.
    1. de Jong TD, Vosslamber S, Blits M, Wolbink G, Nurmohamed MT, van der Laken CJ, Jansen G, Voskuyl AE, Verweij CL. Effect of prednisone on type I interferon signature in rheumatoid arthritis: consequences for response prediction to rituximab. Arthritis Res Ther. 2015;17(1):78. doi: 10.1186/s13075-015-0564-y.
    1. Vosslamber S, Raterman HG, van der Pouw Kraan TC, Schreurs MW, von Blomberg BM, Nurmohamed MT, et al. Pharmacological induction of interferon type I activity following treatment with rituximab determines clinical response in rheumatoid arthritis. Ann Rheum Dis. 2011;70(6):1153–1159. doi: 10.1136/ard.2010.147199.
    1. Llibre A, Bondet V, Rodero MP, Hunt D, Crow YJ, Duffy D. Development and validation of an ultrasensitive single molecule array digital enzyme-linked immunosorbent assay for human interferon-alpha. J Vis Exp. 2018;136.
    1. Mathian A, Mouries-Martin S, Dorgham K, Devilliers H, Yssel H, Garrido Castillo L, Cohen-Aubart F, Haroche J, Hié M, Pineton de Chambrun M, Miyara M, Pha M, Rozenberg F, Gorochov G, Amoura Z. Ultrasensitive serum interferon-α quantification during SLE remission identifies patients at risk for relapse. Ann Rheum Dis. 2019;78(12):1669–1676. doi: 10.1136/annrheumdis-2019-215571.
    1. Smolen JS, Landewé RBM, Bijlsma JWJ, Burmester GR, Dougados M, Kerschbaumer A, McInnes IB, Sepriano A, van Vollenhoven RF, de Wit M, Aletaha D, Aringer M, Askling J, Balsa A, Boers M, den Broeder AA, Buch MH, Buttgereit F, Caporali R, Cardiel MH, de Cock D, Codreanu C, Cutolo M, Edwards CJ, van Eijk-Hustings Y, Emery P, Finckh A, Gossec L, Gottenberg JE, Hetland ML, Huizinga TWJ, Koloumas M, Li Z, Mariette X, Müller-Ladner U, Mysler EF, da Silva JAP, Poór G, Pope JE, Rubbert-Roth A, Ruyssen-Witrand A, Saag KG, Strangfeld A, Takeuchi T, Voshaar M, Westhovens R, van der Heijde D. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2019 update. Ann Rheum Dis. 2020;79(6):685–699. doi: 10.1136/annrheumdis-2019-216655.
    1. Singh JA, Saag KG, Bridges SL, Jr, Akl EA, Bannuru RR, Sullivan MC, et al. 2015 American College of Rheumatology guideline for the treatment of rheumatoid arthritis. Arthritis Care Res. 2016;68(1):1–25. doi: 10.1002/acr.22783.
    1. Hetland ML, Haavardsholm EA, Rudin A, Nordström D, Nurmohamed M, Gudbjornsson B, et al. Active conventional treatment and three different biological treatments in early rheumatoid arthritis: phase IV investigator initiated, randomised, observer blinded clinical trial. BMJ. 2020;371:m4328. doi: 10.1136/bmj.m4328.
    1. Glinatsi D, Heiberg MS, Rudin A, Nordstrom D, Haavardsholm EA, Gudbjornsson B, et al. Head-to-head comparison of aggressive conventional therapy and three biological treatments and comparison of two de-escalation strategies in patients who respond to treatment: study protocol for a multicenter, randomized, open-label, blinded-assessor, phase 4 study. Trials. 2017;18(1):161. doi: 10.1186/s13063-017-1891-x.
    1. Wang C, Kokkonen H, Sandling JK, Johansson M, Seddighzadeh M, Padyukov L, et al. Preferential association of interferon regulatory factor 5 gene variants with seronegative rheumatoid arthritis in 2 Swedish case-control studies. The Journal of Rheumatology. 2011;38(10):2130–2132. doi: 10.3899/jrheum.110322.
    1. Sigurdsson S, Padyukov L, Kurreeman FAS, Liljedahl U, Wiman A-C, Alfredsson L, Toes R, Rönnelid J, Klareskog L, Huizinga TWJ, Alm G, Syvänen AC, Rönnblom L. Association of a haplotype in the promoter region of the interferon regulatory factor 5 gene with rheumatoid arthritis. Arthritis Rheum. 2007;56(7):2202–2210. doi: 10.1002/art.22704.
    1. Sokolove J, Johnson DS, Lahey LJ, Wagner CA, Cheng D, Thiele GM, Michaud K, Sayles H, Reimold AM, Caplan L, Cannon GW, Kerr G, Mikuls TR, Robinson WH. Rheumatoid factor as a potentiator of anti-citrullinated protein antibody-mediated inflammation in rheumatoid arthritis. Arthritis & rheumatology. 2014;66(4):813–821. doi: 10.1002/art.38307.
    1. Ittah M, Miceli-Richard C, Eric Gottenberg J, Lavie F, Lazure T, Ba N, Sellam J, Lepajolec C, Mariette X. B cell-activating factor of the tumor necrosis factor family (BAFF) is expressed under stimulation by interferon in salivary gland epithelial cells in primary Sjögren’s syndrome. Arthritis Res Ther. 2006;8(2):R51. doi: 10.1186/ar1912.
    1. Lundell A-C, Nordström I, Andersson K, Lundqvist C, Telemo E, Nava S, et al. IFN type I and II induce BAFF secretion from human decidual stromal cells. Sci Rep. 2017;7:39904. doi: 10.1038/srep39904.
    1. Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, Banchereau J. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity. 2003;19(2):225–234. doi: 10.1016/S1074-7613(03)00208-5.
    1. Hua J, Kirou K, Lee C, Crow MK. Functional assay of type I interferon in systemic lupus erythematosus plasma and association with anti-RNA binding protein autoantibodies. Arthritis Rheum. 2006;54(6):1906–1916. doi: 10.1002/art.21890.
    1. Furie RA, Morand EF, Bruce IN, Manzi S, Kalunian KC, Vital EM, Lawrence Ford T, Gupta R, Hiepe F, Santiago M, Brohawn PZ, Berglind A, Tummala R. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial. The Lancet Rheumatology. 2019;1(4):e208–ee19. doi: 10.1016/S2665-9913(19)30076-1.
    1. Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, Bae SC, Brohawn PZ, Pineda L, Berglind A, Tummala R. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med. 2020;382(3):211–221. doi: 10.1056/NEJMoa1912196.
    1. Reynier F, Petit F, Paye M, Turrel-Davin F, Imbert PE, Hot A, Mougin B, Miossec P. Importance of correlation between gene expression levels: application to the type I interferon signature in rheumatoid arthritis. PLoS One. 2011;6(10):e24828. doi: 10.1371/journal.pone.0024828.
    1. Mavragani CP, La DT, Stohl W, Crow MK. Association of the response to tumor necrosis factor antagonists with plasma type I interferon activity and interferon-beta/alpha ratios in rheumatoid arthritis patients: a post hoc analysis of a predominantly Hispanic cohort. Arthritis Rheum. 2010;62(2):392–401. doi: 10.1002/art.27226.
    1. van der Pouw Kraan TC, Wijbrandts CA, van Baarsen LG, Voskuyl AE, Rustenburg F, Baggen JM, et al. Rheumatoid arthritis subtypes identified by genomic profiling of peripheral blood cells: assignment of a type I interferon signature in a subpopulation of patients. Ann Rheum Dis. 2007;66(8):1008–1014. doi: 10.1136/ard.2006.063412.
    1. de Jong TD, Snoek T, Mantel E, van der Laken CJ, van Vollenhoven RF, Lems WF. Dynamics of the type I interferon response during immunosuppressive therapy in rheumatoid arthritis. Front Immunol. 2019;10:902. doi: 10.3389/fimmu.2019.00902.

Source: PubMed

3
Sottoscrivi