Safety and efficacy of filgotinib, lanraplenib and tirabrutinib in Sjögren's syndrome: a randomized, phase 2, double-blind, placebo-controlled study

Elizabeth Price, Michele Bombardieri, Alan Kivitz, Franziska Matzkies, Oksana Gurtovaya, Alena Pechonkina, Wendy Jiang, Bryan Downie, Anubhav Mathur, Afsaneh Mozaffarian, Neelufar Mozaffarian, J Eric Gottenberg, Elizabeth Price, Michele Bombardieri, Alan Kivitz, Franziska Matzkies, Oksana Gurtovaya, Alena Pechonkina, Wendy Jiang, Bryan Downie, Anubhav Mathur, Afsaneh Mozaffarian, Neelufar Mozaffarian, J Eric Gottenberg

Abstract

Objective: The aim of this study was to characterize the safety and efficacy of filgotinib, lanraplenib and tirabrutinib in patients with active SS.

Methods: This multicentre, double-blind study randomized patients with active primary or secondary SS [EULAR SS disease activity index (ESSDAI) ≥5) to receive filgotinib 200 mg (Janus kinase-1 inhibitor), lanraplenib 30 mg (spleen tyrosine kinase inhibitor), tirabrutinib 40 mg (Bruton's tyrosine kinase inhibitor), or placebo. The composite primary end point was the week-12 proportion of patients fulfilling protocol-specified improvement criteria (based on CRP and SS-related symptoms). The EULAR SS patient-reported index (ESSPRI) and the ESSDAI change from baseline (CFB) were secondary end points. Exploratory end points included disease-related biomarkers. Treatment-emergent adverse events (AEs) represented safety outcomes.

Results: The mean of the baseline ESSDAI was 10.1, and of ESSPRI was 6.2 in the 150 patients who were treated; 125 completed the 24-week placebo-controlled treatment period. At week 12, 43.3% of the filgotinib group achieved the primary end point (P = 0.17 vs placebo) vs 42.3% (P = 0.16), 34.7% (P = 0.33), and 26.7% of lanraplenib, tirabrutinib, and placebo groups, respectively. Neither secondary end point was met. Biomarker reductions included immunoglobulins classically associated with SS disease activity. Filgotinib ESSDAI CFB appeared more pronounced in subgroups with baseline ESSDAI ≥14 or without DMARDs/CSs. Most AEs were Grade 1 or 2.

Conclusion: Three drugs with disparate mechanisms were tested, but no significant differences vs placebo in primary or secondary end points were observed. These results may be considered hypothesis-generating, given the drug tolerability, subgroup analysis, and biomarker findings.

Trial registration: ClinicalTrials.gov, https://ichgcp.net/clinical-trials-registry/NCT03100942" title="See in ClinicalTrials.gov">NCT03100942.

Keywords: Sjögren's syndrome; efficacy; filgotinib; lanraplenib; randomized trial; safety; tirabrutinib.

© The Author(s) 2022. Published by Oxford University Press on behalf of the British Society for Rheumatology.

Figures

Fig . 1
Fig. 1
Study design and disposition (A) Study schema. After screening, eligible patients were randomized 1:1:1:1 in a blinded fashion to receive FIL 200 mg QD (n = 38), LANRA 30 mg QD (n = 37), TIRA 40 mg QD (n = 39) or PBO (n = 36) for 24 weeks. Initial randomization was stratified by concurrent use of systemic CSs or csDMARDs and by ESSDAI haematologic + biological domain score <2 or ≥2. The primary end point was assessed at W12 and secondary end points at W12 and W24. The FIL, LANRA and TIRA groups continued treatment until W48. At W24, the PBO group was re-randomized 1:1:1 into other treatment groups until W48. All patients had 4-week follow-up at W52. (B) Study disposition by treatment phase. One hundred and fifty-two patients were randomized; 150 patients received at least one dose of a study drug and were included in the safety analysis set. Thirty-four (89.5%) and 30 (78.9%) of the patients in the FIL group completed treatment at W12 and W24, respectively. Thirty (81.1%) and 27 (73.0%) of the patients in the LANRA group completed treatment at W12 and W24, respectively. Thirty-six (92.3%) of the patients in the TIRA group completed treatment at W12 and W24. Thirty-four (94.4%) and 32 (88.9%) of the patients in the PBO group completed treatment at W12 and W24, respectively. BIO: biological component score of ESSDAI; csDMARD: conventional synthetic DMARD; ESSDAI: EULAR SS disease activity index; FIL: filgotinib; HEM: haematologic component score of ESSDAI; LANRA: lanraplenib; PBO: placebo; QD: once daily; TIRA: tirabrutinib; W: week.
Fig . 2
Fig. 2
Primary end point, proportion of responders at W12 The primary end point is summarized using the full analysis set, which includes patients who were randomized and received at least one dose of the study drug. Error bars show 95% CIs. Numbers on graph show difference in response rate vs PBO, 95% CI of differences vs placebo, and the P-value of the active treatment group compared with PBO. FIL: filgotinib; LANRA: lanraplenib; PBO: placebo; TIRA: tirabrutinib; W: week.
Fig . 3
Fig. 3
ESSDAI score–adjusted mean change from baseline (A) Overall study population. Error bars show 95% CI of LS mean. LS means, 95% CIs and P values were obtained from a mixed-effects model for repeated measures with the terms for baseline value, treatment, stratification factors, visit, and treatment-by-visit interaction. **P < 0.01 vs placebo. The baseline value was the last available value collected on or prior to first dose of the study drug (day 1). (B) ESSDAI score–adjusted mean change from baseline in patients with baseline ESSDAI ≥14. Markers indicate median; error bars indicate interquartile range. Numbers below the graph show numbers of patients. The baseline value was the last available value collected on or prior to the first dose of the study drug (day 1). (C) The ESSDAI score–adjusted mean change from baseline in patients not taking DMARDs/CSs at baseline. Markers indicate median; error bars indicate interquartile range. Numbers below the graph show numbers of patients. The baseline value was the last available value collected on or prior to the first dose of the study drug (day 1). ESSDAI: EULAR SS disease activity index; FIL: filgotinib; LANRA: lanraplenib; LS: least square; PBO: placebo; Q1: first quartile; Q3: third quartile; TIRA: tirabrutinib.
Fig . 4
Fig. 4
Biomarker changes (A) Median percentage change in biomarker activity after treatment. The heat map represents changes from baseline at Weeks 4, 12 and 24 (bottom labels) in each treatment group (top labels). At baseline, 50%, 71.4%, 52.6% and 71.4% of patients in the filgotinib, lanraplenib, tirabrutinib and placebo groups had RF values below the LLOQ; 2.6% of patients in the tirabrutinib group had IgM levels below LLOQ; no patients had IgG levels below LLOQ; and 8.3% of patients in the placebo group had CRP levels below LLOQ. Treated groups were compared with placebo by the Wilcoxon rank-sum test with Hommel’s method for multiplicity adjustment. ***P < 0.001, **P < 0.01, *P < 0.05. (B) Change in Type I IFN signature from baseline by treatment group. The y-axis shows the adjusted mean and 95% CI of change in the pathway activity score following treatment. The outlined circle is a statistically significant difference compared with baseline and placebo. LLOQ: lower limit of quantification.

References

    1. Patel R, Shahane A.. The epidemiology of Sjögren’s syndrome. Clin Epidemiol 2014;6:247–55.
    1. Miyamoto ST, Valim V, Fisher BA.. Health-related quality of life and costs in Sjögren’s syndrome. Rheumatology (Oxford) 2021;60:2588–601.
    1. Retamozo S, Acar-Denizli N, Rasmussen A. et al. Systemic manifestations of primary Sjögren’s syndrome out of the ESSDAI classification: prevalence and clinical relevance in a large international, multi-ethnic cohort of patients. Clin Exp Rheumatol 2019;37(Suppl 118):97–106.
    1. Vivino FB, Bunya VY, Massaro-Giordano G. et al. Sjogren’s syndrome: an update on disease pathogenesis, clinical manifestations and treatment. Clin Immunol 2019;203:81–121.
    1. Felten R, Scher F, Sibilia J, Gottenberg JE, Arnaud L.. The pipeline of targeted therapies under clinical development for primary Sjögren’s syndrome: a systematic review of trials. Autoimmun Rev 2019;18:576–82.
    1. Skopouli FN, Jagiello P, Tsifetaki N, Moutsopoulos HM.. Methotrexate in primary Sjögren’s syndrome. Clin Exp Rheumatol 1996;14:555–8.
    1. Devauchelle-Pensec V, Mariette X, Jousse-Joulin S. et al. Treatment of primary Sjögren syndrome with rituximab: a randomized trial. Ann Intern Med 2014;160:233–42.
    1. Bowman SJ, Everett CC, O’Dwyer JL. et al. Randomized controlled trial of rituximab and cost-effectiveness analysis in treating fatigue and oral dryness in primary Sjögren’s syndrome. Arthritis Rheumatol 2017;69:1440–50.
    1. Meijer JM, Meiners PM, Vissink A. et al. Effectiveness of rituximab treatment in primary Sjögren’s syndrome: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum 2010;62:960–8.
    1. Demarchi J, Papasidero S, Medina MA. et al. Primary Sjögren’s syndrome: extraglandular manifestations and hydroxychloroquine therapy. Clin Rheumatol 2017;36:2455–60.
    1. Fisher BS, Ng W-F, Bombardieri M. et al. Assessment of the anti-CD40 antibody iscalimab in patients with primary Sjögren’s syndrome: a multicentre, randomised, double-blind, placebo-controlled, proof-of-concept study. Lancet Rheumatol 2020;2:e142–52.
    1. Baer AN, Gottenberg JE, St Clair EW. et al. Efficacy and safety of abatacept in active primary Sjögren’s syndrome: results of a phase III, randomised, placebo-controlled trial. Ann Rheum Dis 2021;80:339–48.
    1. van Nimwegen JF, Mossel M, van Zuiden GS. et al. Abatacept treatment for patients with early active primary Sjögren’s syndrome: a single-centre, randomised, double-blind, placebo-controlled, phase 3 trial (ASAP-III study). Lancet Rheumatology 2020;2:e153–63.
    1. Ramos-Casals M, Brito-Zeron P, Bombardieri S. et al.; EULAR-Sjögren Syndrome Task Force Group. EULAR recommendations for the management of Sjögren’s syndrome with topical and systemic therapies. Ann Rheum Dis 2020;79:3–18.
    1. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM.. JAK-STAT signaling as a target for inflammatory and autoimmune diseases: current and future prospects. Drugs 2017;77:521–46.
    1. Roescher N, Tak PP, Illei GG.. Cytokines in Sjögren’s syndrome. Oral Dis 2009;15:519–26.
    1. Corneth OBJ, Verstappen GMP, Paulissen SMJ. et al. Enhanced Bruton’s tyrosine kinase activity in peripheral blood B lymphocytes from patients with autoimmune disease. Arthritis Rheumatol 2017;69:1313–24.
    1. Imgenberg-Kreuz J, Sandling JK, Bjork A. et al. Transcription profiling of peripheral B cells in antibody-positive primary Sjögren’s syndrome reveals upregulated expression of CX3CR1 and a type I and type II interferon signature. Scand J Immunol 2018;87:e12662.
    1. Fox RI, Fox CM, Gottenberg JE, Dorner T.. Treatment of Sjögren’s syndrome: current therapy and future directions. Rheumatology (Oxford) 2021;60:2066–74.
    1. Jyseleca® (Filgotinib maleate tablets). Japanese Package Insert. Gilead Sciences K.K. Tokyo, Japan. 2020. Available at: (5 April 2022, date last accessed).
    1. Jyseleca® (filgotinib) 100 and 200 mg film-coated tablets. Summary of Product Characteristics. Gilead Sciences Ltd. London, UK. 2020. (5 April 2022, date last accessed).
    1. Blomgren P, Chandrasekhar J, Di Paolo JA. et al. Discovery of Lanraplenib (GS-9876): a once-daily spleen tyrosine kinase inhibitor for autoimmune diseases. ACS Med Chem Letters 2020;11:506–13.
    1. Dhillon S. Tirabrutinib: first approval. Drugs 2020;80:835–40.
    1. Bahjat FR, Pine PR, Reitsma A. et al. An orally bioavailable spleen tyrosine kinase inhibitor delays disease progression and prolongs survival in murine lupus. Arthritis Rheum 2008;58:1433–44.
    1. Hutcheson J, Vanarsa K, Bashmakov A. et al. Modulating proximal cell signaling by targeting Btk ameliorates humoral autoimmunity and end-organ disease in murine lupus. Arthritis Res Ther 2012;14:R243–R.
    1. Rankin AL, Seth N, Keegan S. et al. Selective inhibition of BTK prevents murine lupus and antibody-mediated glomerulonephritis. J Immunol 2013;191:4540–50.
    1. Seror R, Bootsma H, Saraux A. et al. Defining disease activity states and clinically meaningful improvement in primary Sjögren’s syndrome with EULAR primary Sjögren’s syndrome disease activity (ESSDAI) and patient-reported indexes (ESSPRI). Ann Rheum Dis 2016;75:382–9.
    1. van der Heijden EB, Hillen MR, Lopes APP. et al. Leflunomide–hydroxychloroquine combination therapy in patients with primary Sjögren’s syndrome (RepurpSS-I): a placebo-controlled, double-blinded, randomised clinical trial. Lancet Rheumatol 2020;2:e260–9.
    1. Seror R, Bowman SJ, Brito-Zeron P. et al. EULAR Sjögren’s syndrome disease activity index (ESSDAI): a user guide. RMD Open 2015;1:e000022.
    1. Seror R, Ravaud P, Mariette X. et al. 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. Avina-Zubieta JA, Jansz M, Sayre EC, Choi HK.. The risk of deep venous thrombosis and pulmonary embolism in primary Sjögren syndrome: a general population-based study. J Rheumatol 2017;44:1184–9.
    1. Nash P, Kerschbaumer A, Dorner T. et al. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis 2021;80:71–87.
    1. Bazzan M, Vaccarino A, Marletto F.. Systemic lupus erythematosus and thrombosis. Thromb J 2015;13:16.
    1. Rigel Pharmaceuticals Inc. TAVALISSE® (fostamatinib disodium hexahydrate) tablets [prescribing information]. 2020. (5 April 2022, date last accessed).
    1. Pharmacyclics LLC. IMBRUVICA® (ibrutinib) [prescribing information]. 2020. (5 April 2022, date last accessed).
    1. de Wolff L, Arends S, van Nimwegen JF, Bootsma H.. Ten years of the ESSDAI: is it fit for purpose? Clin Exp Rheumatol 2020;38(Suppl 126):283–90.
    1. Felten R, Gottenberg JE.. Advances in treatments for Sjögren’s syndrome: the glass is half full. Lancet Rheumatology 2020;2:e516–8.
    1. Benedetti F, Carlino E, Pollo A.. How placebos change the patient’s brain. Neuropsychopharmacology 2011;36:339–54.
    1. Arends S. D, van Nimwegen JG, Verstappen GMPJ. et al. Composite of Relevant Endpoints for Sjögren’s Syndrome (CRESS): development and validation of a novel outcome measure. Lancet Rheumatol 2021;3:E553–62.
    1. Narváez J, Sánchez-Fernández SÁ, Seoane-Mato D, Díaz-González F, Bustabad S.. Prevalence of Sjögren’s syndrome in the general adult population in Spain: estimating the proportion of undiagnosed cases. Sci Rep 2020;10:10627.

Source: PubMed

3
Abonneren