9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      High levels of circulating interferons type I, type II and type III associate with distinct clinical features of active systemic lupus erythematosus

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background and aim

          Interferons (IFNs) are considered to be key molecules in the pathogenesis of systemic lupus erythematosus (SLE). We measured levels of type I, II and III IFNs in a large cohort of patients with systemic lupus erythematosus (SLE) and controls and explored associations among high levels of different IFN types and distinct SLE features.

          Methods

          Four hundred ninety-seven well-characterized SLE patients and 322 population controls were included. Disease activity was assessed by SLE Disease Activity Index (SLEDAI) and Systemic Lupus Activity Measure (SLAM). Functional type I IFN activity was estimated by a WISH reporter cell assay. Levels of IFN-γ were estimated by MSD 30-plex assay. IFN-α and IFN-λ1 were measured by ELISA. Values above the third quartile of patients’ measurements were defined as high. Associations among high IFN results and SLE features were investigated by nominal regression analysis.

          Results

          All IFN measurements were higher in SLE patients than in controls. High type I IFN activity correlated with levels of IFN-γ and IFN-α and associated with active SLE in most domains: weight loss, fatigue, fever, rash, lymphadenopathy, arthritis, nephritis and haematological manifestations. Specific SLE subsets were linked to the upregulation of different subtypes of circulating IFNs: high IFN-γ to arthritis, nephritis and anti-Ro60 antibodies and high IFN-α to mucocutaneous engagement and anti-Ro52 and anti-La antibodies. Isolated high IFN-λ1 was coupled to anti-nucleosome antibodies and less severe SLE.

          Conclusions

          High functional type I IFN activity captures active SLE in most domains, but more distinct patterns of organ involvement are associated with profiles of circulating IFNs. High IFN-γ as well as high functional type I IFN activity is a characteristic of severe SLE with nephritis and arthritis, while elevated levels of IFN-α associate with active mucocutaneous inflammation and a more benign cardiovascular profile. IFN-λ1 in isolation is associated with milder disease. Our findings suggest that IFNs contribute to the heterogeneity of clinical manifestations in SLE, and measuring circulating IFNs could assist in designing clinical trials with therapies targeting IFN pathways.

          Electronic supplementary material

          The online version of this article (10.1186/s13075-019-1878-y) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references29

          • Record: found
          • Abstract: found
          • Article: not found

          Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus.

          Systemic lupus erythematosus (SLE) is a complex, inflammatory autoimmune disease that affects multiple organ systems. We used global gene expression profiling of peripheral blood mononuclear cells to identify distinct patterns of gene expression that distinguish most SLE patients from healthy controls. Strikingly, about half of the patients studied showed dysregulated expression of genes in the IFN pathway. Furthermore, this IFN gene expression "signature" served as a marker for more severe disease involving the kidneys, hematopoetic cells, and/or the central nervous system. These results provide insights into the genetic pathways underlying SLE, and identify a subgroup of patients who may benefit from therapies targeting the IFN pathway.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Anifrolumab, an Anti–Interferon‐α Receptor Monoclonal Antibody, in Moderate‐to‐Severe Systemic Lupus Erythematosus

            Objective To assess the efficacy and safety of anifrolumab, a type I interferon (IFN) receptor antagonist, in a phase IIb, randomized, double‐blind, placebo‐controlled study of adults with moderate‐to‐severe systemic lupus erythematosus (SLE). Methods Patients (n = 305) were randomized to receive intravenous anifrolumab (300 mg or 1,000 mg) or placebo, in addition to standard therapy, every 4 weeks for 48 weeks. Randomization was stratified by SLE Disease Activity Index 2000 score (<10 or ≥10), oral corticosteroid dosage (<10 or ≥10 mg/day), and type I IFN gene signature test status (high or low) based on a 4‐gene expression assay. The primary end point was the percentage of patients achieving an SLE Responder Index (SRI[4]) response at week 24 with sustained reduction of oral corticosteroids (<10 mg/day and less than or equal to the dose at week 1 from week 12 through 24). Other end points (including SRI[4], British Isles Lupus Assessment Group [BILAG]–based Composite Lupus Assessment [BICLA], modified SRI[6], and major clinical response) were assessed at week 52. The primary end point was analyzed in the modified intent‐to‐treat (ITT) population and type I IFN–high subpopulation. The study result was considered positive if the primary end point was met in either of the 2 study populations. The Type I error rate was controlled at 0.10 (2‐sided), within each of the 2 study populations for the primary end point analysis. Results The primary end point was met by more patients treated with anifrolumab (34.3% of 99 for 300 mg and 28.8% of 104 for 1,000 mg) than placebo (17.6% of 102) (P = 0.014 for 300 mg and P = 0.063 for 1,000 mg, versus placebo), with greater effect size in patients with a high IFN signature at baseline (13.2% in placebo‐treated patients versus 36.0% [P = 0.004] and 28.2% [P = 0.029]) in patients treated with anifrolumab 300 mg and 1,000 mg, respectively. At week 52, patients treated with anifrolumab achieved greater responses in SRI(4) (40.2% versus 62.6% [P < 0.001] and 53.8% [P = 0.043] with placebo, anifrolumab 300 mg, and anifrolumab 1,000 mg, respectively), BICLA (25.7% versus 53.5% [P < 0.001] and 41.2% [P = 0.018], respectively), modified SRI(6) (28.4% versus 49.5% [P = 0.002] and 44.7% [P = 0.015], respectively), major clinical response (BILAG 2004 C or better in all organ domains from week 24 through week 52) (6.9% versus 19.2% [P = 0.012] and 17.3% [P = 0.025], respectively), and several other global and organ‐specific end points. Herpes zoster was more frequent in the anifrolumab‐treated patients (2.0% with placebo treatment versus 5.1% and 9.5% with anifrolumab 300 mg and 1,000 mg, respectively), as were cases reported as influenza (2.0% versus 6.1% and 7.6%, respectively), in the anifrolumab treatment groups. Incidence of serious adverse events was similar between groups (18.8% versus 16.2% and 17.1%, respectively). Conclusion Anifrolumab substantially reduced disease activity compared with placebo across multiple clinical end points in the patients with moderate‐to‐severe SLE.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease.

              Gene-expression studies have demonstrated increased expression of interferon (IFN)-inducible genes (IFIGs) in peripheral blood mononuclear cells (PBMCs) of many patients with systemic lupus erythematosus (SLE), with a predominant effect of type I IFN. This study examined the hypothesis that increased disease severity and activity, as well as distinct autoantibody specificities, characterize SLE patients with activation of the type I IFN pathway. Freshly isolated PBMCs from 77 SLE patients, 22 disease controls, and 28 healthy donors were subjected to real-time polymerase chain reaction for 3 IFIGs that are preferentially induced by IFNalpha, and the data were used to derive IFNalpha scores for all individuals. Expression of IFIGs was significantly higher in SLE patients compared with disease controls or healthy donors. SLE patients with high and low IFNalpha scores were compared for clinical manifestations of disease, disease severity, disease activity, serologic features, and potential confounders, by bivariate and multivariate analyses. SLE patients with a high IFNalpha score had a significantly higher prevalence of renal disease, a greater number of American College of Rheumatology criteria for SLE, and a higher Systemic Lupus International Collaborating Clinics damage index (SDI) score than did SLE patients with low IFNalpha scores. Patients with high scores showed increased disease activity, as measured by lower C3 levels, hemoglobin levels, absolute lymphocyte counts, and albumin levels, and a higher anti-double-stranded DNA (dsDNA) titer, erythrocyte sedimentation rate, and SLE Disease Activity Index 2000 score. The presence of antibodies specific for Ro, U1 RNP, Sm, and dsDNA, but not phospholipids, was significantly associated with a high IFNalpha score. Logistic regression analysis confirmed that renal disease, higher SDI scores, low complement levels, and presence of anti-RNA binding protein (RBP) autoantibodies were associated with a high IFNalpha score. Activation of the IFNalpha pathway defines a subgroup of SLE patients whose condition is characterized by increased disease severity, including renal disease, increased disease activity, reflected in complement activation, and autoreactivity to RBP.
                Bookmark

                Author and article information

                Contributors
                +46 72 452 1290 , vilija.oke@ki.se
                iva.gunnarsson@sll.se
                Dorschner.Jessica@mayo.edu
                Susanna.Eketjall@astrazeneca.com
                agneta.zickert@sll.se
                Timothy.Niewold@nyumc.org
                elisabet.svenungsson@ki.se
                Journal
                Arthritis Res Ther
                Arthritis Res. Ther
                Arthritis Research & Therapy
                BioMed Central (London )
                1478-6354
                1478-6362
                29 April 2019
                29 April 2019
                2019
                : 21
                : 107
                Affiliations
                [1 ]ISNI 0000 0000 9241 5705, GRID grid.24381.3c, Rheumatology Unit, Department of Medicine, Karolinska Institutet, , Karolinska University Hospital, ; SE-171 76 Stockholm, Sweden
                [2 ]ISNI 0000 0004 0459 167X, GRID grid.66875.3a, Division of Rheumatology and Department of Immunology, , Mayo Clinic, ; Rochester, MN USA
                [3 ]ISNI 0000 0004 1937 0626, GRID grid.4714.6, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Integrated Cardio Metabolic Centre, , Karolinska Institutet, ; Huddinge, Sweden
                [4 ]ISNI 0000 0004 1936 8753, GRID grid.137628.9, Colton Center for Autoimmunity, , New York University School of Medicine, ; New York, NY USA
                Author information
                http://orcid.org/0000-0002-1834-2688
                Article
                1878
                10.1186/s13075-019-1878-y
                6489203
                31036046
                56017034-75e3-4d71-b9cb-36837441c8f5
                © The Author(s). 2019

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 22 January 2019
                : 26 March 2019
                Funding
                Funded by: Karolinska Institutet - Mayo Clinic collaboration grant
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100003793, Hjärt-Lungfonden;
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004359, Vetenskapsrådet;
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004348, Stockholms Läns Landsting;
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100007857, Stiftelsen Konung Gustaf V:s 80-årsfond;
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100007949, Reumatikerförbundet;
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100007687, Svenska Läkaresällskapet;
                Award ID: Not applicable
                Award Recipient :
                Funded by: Ingegerd Johansson´s foundation
                Award ID: Not applicable
                Award Recipient :
                Funded by: The Fund for Renal research
                Award ID: Not applicable
                Award Recipient :
                Funded by: Signe and Reinhold Sunds foundation
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100004047, Karolinska Institutet;
                Award ID: Not applicable
                Award Recipient :
                Funded by: Colton Center for Autoimmunity
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000009, Foundation for the National Institutes of Health;
                Award ID: AR060861, AR057781, AR065964, AI071651
                Award Recipient :
                Funded by: Rheumatology Research Foundation,
                Award ID: Not applicable
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000871, Mayo Clinic;
                Award ID: Not applicable
                Award Recipient :
                Funded by: the Lupus Research Foundation
                Award ID: Not applicable
                Award Recipient :
                Categories
                Research Article
                Custom metadata
                © The Author(s) 2019

                Orthopedics
                sle,interferons,disease activity,autoantibodies
                Orthopedics
                sle, interferons, disease activity, autoantibodies

                Comments

                Comment on this article