Improvement of Severe Fatigue Following Nuclease Therapy in Patients With Primary Sjögren's Syndrome: A Randomized Clinical Trial

James Posada, Saba Valadkhan, Daniel Burge, Kristen Davies, Jessica Tarn, John Casement, Kerry Jobling, Peter Gallagher, Douglas Wilson, Francesca Barone, Benjamin A Fisher, Wan-Fai Ng, James Posada, Saba Valadkhan, Daniel Burge, Kristen Davies, Jessica Tarn, John Casement, Kerry Jobling, Peter Gallagher, Douglas Wilson, Francesca Barone, Benjamin A Fisher, Wan-Fai Ng

Abstract

Objective: To assess the safety and efficacy of RSLV-132, an RNase Fc fusion protein, in a phase II randomized, double-blind, placebo-controlled clinical trial in patients with primary Sjögren's syndrome (SS).

Methods: Thirty patients with primary SS were randomized to receive treatment with RSLV-132 or placebo intravenously once per week for 2 weeks, and then every 2 weeks for 12 weeks. Eight patients received placebo and 20 patients received RSLV-132 at a dose of 10 mg/kg. Clinical efficacy measures included the European League Against Rheumatism (EULAR) Sjögren's Syndrome Disease Activity Index, EULAR Sjögren's Syndrome Patient Reported Index (ESSPRI), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F), Profile of Fatigue (ProF), and the Digit Symbol Substitution Test (DSST).

Results: Patients randomized to receive RSLV-132 experienced clinically meaningful improvements in the ESSPRI score (P = 0.27), FACIT-F score (P = 0.05), ProF score (P = 0.07), and DSST (P = 0.02) from baseline to day 99, whereas patients who received placebo showed no changes in any of these clinical efficacy measures. This improvement was significantly correlated with increased expression of selected interferon-inducible genes (Pearson's correlations, each P < 0.05).

Conclusion: Administration of RSLV-132 improved severe fatigue, as determined by 4 independent patient-reported measures of fatigue, in patients with primary SS.

Trial registration: ClinicalTrials.gov NCT03247686.

© 2020 Resolve Therapeutics, LLC. Arthritis & Rheumatology published by Wiley Periodicals, Inc. on behalf of American College of Rheumatology.

Figures

Figure 1
Figure 1
Distribution of patients with primary Sjögren’s syndrome to the randomized treatment groups, with follow‐up. Two subjects did not meet the eligibility criterion requiring serum positivity for anti‐Ro autoantibodies. Eligible patients were randomized to receive RSLV‐132 (10 mg/kg) or placebo, once weekly for 2 weeks and then every 2 weeks for 12 weeks.
Figure 2
Figure 2
Heatmaps showing changes in expression of interferon (IFN)–inducible genes from day 1 to day 99. The log2 fold change in expression of 3 modules (M1.2, M3.4, and M5.12) of IFN‐inducible genes was assessed in whole blood samples from the placebo group (n = 7) compared to the RSLV‐132 group as a whole (n = 20) (A) or the subgroups of RSLV‐132–treated patients who either achieved a clinical response (R) or did not achieve a clinical response (NR) (B) over the follow‐up.
Figure 3
Figure 3
Secondary end point efficacy measures. Clinical efficacy was assessed as the mean change from baseline in the fatigue component of the European League Against Rheumatism Sjögren’s Syndrome Patient Reported Index (ESSPRI) (A), Functional Assessment of Chronic Illness Therapy–Fatigue (FACIT‐F) (B), and mental fatigue component of the Profile of Fatigue (ProF) (C) in the RSLV‐132 and placebo treatment groups. Groups were compared using separate 1‐way analysis of variance models for each visit, each testing the null hypothesis that the true mean difference between treatment groups was 0 (unadjusted α = 0.05). Between‐group differences were as follows: P = 0.136 in A, P = 0.092 in B, and P = 0.046 in C. Results at each time point are the mean ± SEM.

References

    1. Haldorsen K, Bjelland I, Bolstad AI, Jonsson R, Brun JG. A five‐year prospective study of fatigue in primary Sjögren’s syndrome. Arthritis Res Ther 2011;13:R167.
    1. Overman CL, Kool MB, da Silva JA, Geenen R. The prevalence of severe fatigue in rheumatic diseases: an international study. Clin Rheumatol 2016;35:409–15.
    1. Bodewes IL, van der Spek PJ, Leon LG, Wijkhuijs AJ, van Helden‐Meeuwsen CG, Tas L, et al. Fatigue in Sjögren’s syndrome: a search for biomarkers and treatment targets. Front Immunol 2019;10:312.
    1. Nezos A, Gravani F, Tassidou A, Kapsogeorgou EK, Voulgarelis M, Koutsilieris M, et al. Type I and II interferon signatures in Sjögren’s syndrome pathogenesis: contributions in distinct clinical phenotypes and Sjögren’s related lymphomagenesis. J Autoimmun 2015;63:47–58.
    1. Hall JC, Baer AN, Shah AA, Criswell LA, Shiboski CH, Rosen A, et al. Molecular subsetting of interferon pathways in Sjögren’s syndrome. Arthritis Rheumatol 2015;67:2437–46.
    1. Driedonks TA, Hoen EN. Circulating Y‐RNAs in extracellular vesicles and ribonucleoprotein complexes: implications for the immune system [review]. Front Immunol 2019;9:3164.
    1. Katze MG, Fornek JL, Palermo RE, Walters KA, Korth MJ. Innate immune modulation by RNA viruses: emerging insights from functional genomics [review]. Nat Rev Immunol 2008;8:644–54.
    1. Moutsopoulos HM, Zerva LV. Anti‐Ro (SSA)/La (SSB) antibodies and Sjögren’s syndrome. Clin Rheumatol 1990;9:123–30.
    1. Kattah NH, Kattah MG, Utz PJ. The U1‐snRNP complex: structural properties relating to autoimmune pathogenesis in rheumatic diseases [review]. Immunol Rev 2010;233:126–45.
    1. Noll F, Behnke J, Leiting S, Troidl K, Alves GT, Müller‐Redetzky H, et al. Self‐extracellular RNA acts in synergy with exogenous danger signals to promote inflammation. PLoS One 2017;12:e0190002.
    1. Eloranta ML, Lövgren T, Finke D, Mathsson L, Rönnelid J, Kastner B, et al. Regulation of the interferon‐α production induced by RNA‐containing immune complexes in plasmacytoid dendritic cells. Arthritis Rheum 2009;60:2418–27.
    1. Doedens JR, Jones WD, Hill K, Mason MJ, Gersuk VH, Mease PJ, et al. Blood‐borne RNA correlates with disease activity and IFN‐stimulated gene expression in systemic lupus erythematosus. J Immunol 2016;197:2854–63.
    1. Hur K, Kim SH, Kim JM. Potential implications of long noncoding RNAs in autoimmune diseases [review]. Immune Netw 2019;19:e4.
    1. Burge DJ, Eisenman J, Byrnes‐Blake K, Smolak P, Lau K, Cohen SB, et al. Safety, pharmacokinetics, and pharmacodynamics of RSLV‐132, an RNase‐Fc fusion protein in systemic lupus erythematosus: a randomized, double‐blind, placebo‐controlled study. Lupus 2017;26:825–34.
    1. Chiche L, Jourde‐Chiche N, Whalen E, Presnell S, Gersuk V, Dang K, et al. Modular transcriptional repertoire analyses of adults with systemic lupus erythematosus reveal distinct type I and type II interferon signatures. Arthritis Rheumatol 2014;66:1583–95.
    1. Seror R, Ravaud P, Bowman SJ, Baron G, Tzioufas A, Theander E, et al, on behalf of the EULAR Sjögren’s Task Force . EULAR Sjögren’s Syndrome Disease Activity Index: development of a consensus systemic disease activity index for primary Sjögren’s syndrome. Ann Rheum Dis 2010;69:1103–9.
    1. Seror R, Ravaud P, Mariette X, Bootsma H, Theander E, Hansen A, et al, on behalf of the EULAR Sjögren’s Task Force . EULAR Sjögren’s Syndrome Patient Reported Index (ESSPRI): development of a consensus patient index for primary Sjögren’s syndrome. Ann Rheum Dis 2011;70:968–72.
    1. Cella D, Yount S, Sorensen M, Chartash E, Sengupta N, Grober J. Validation of the Functional Assessment of Chronic Illness Therapy Fatigue Scale relative to other instrumentation in patients with rheumatoid arthritis. J Rheumatol 2005;32:811–9.
    1. Bowman SJ, Booth DA, Platts RG, UK Sjögren’s Interest Group . Measurement of fatigue and discomfort in primary Sjögren’s syndrome using a new questionnaire tool. Rheumatology (Oxford) 2004;43:758–64.
    1. Wechsler D. Wechsler Adult Intelligence Scale‐Revised. San Antonio, TX: The Psychological Corporation; 1991.
    1. Bray NL, Pimentel H, Melsted P, Pachter L. Near‐optimal probabilistic RNA‐seq quantification. Nat Biotechnol 2016;34:525–7.
    1. Tripp HN, Tarn J, Natasari A, Gillespie C, Mitchell S, Hackett KL, et al. Fatigue in primary Sjögren’s syndrome is associated with lower levels of proinflammatory cytokines. RMD Open 2016;2:e000282.
    1. Davies K, Mirza K, Tarn J, Howard‐Tripp N, Bowman SJ, Lendrem D, et al. Fatigue in primary Sjögren’s syndrome (pSS) is associated with lower levels of proinflammatory cytokines: a validation study. Rheumatol Int 2019;39:1867–73.
    1. Bodewes IL, Al‐Ali S, van Helden‐Meeuwsen CG, Maria NI, Tarn J, Lendrem DW, et al. Systemic interferon type I and type II signatures in primary Sjögren’s syndrome reveal differences in biological disease activity. Rheumatology (Oxford) 2018;57:921–30.
    1. Tarn JR, Howard‐Tripp N, Lendrem DW, Mariette X, Saraux A, Devauchelle‐Pensec V, et al. Symptom‐based stratification of patients with primary Sjögren’s syndrome: multi‐dimensional characterisation of international observational cohorts and reanalyses of randomised clinical trials. Lancet Rheumatol 2019;1:e85–94.
    1. Bodewes IL, Gottenberg JE, van Helden‐Meeuwsen CG, Mariette X, Versnel MA. Hydroxychloroquine treatment downregulates systemic interferon activation in primary Sjögren’s syndrome in the JOQUER randomized trial. Rheumatology (Oxford) 2020;59:107–11.
    1. Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez C, et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med 2020;382:211–21.
    1. Arroyo JD, Chevillet JR, Kroh EM, Ruf IK, Pritchard CC, Givson DF, et al. Argonaut‐2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc Natl Acad Sci U S A 2011;108:5003–8.

Source: PubMed

3
Iratkozz fel