Filgotinib or lanraplenib in moderate to severe cutaneous lupus erythematosus: a phase 2, randomized, double-blind, placebo-controlled study

Victoria P Werth, Roy Fleischmann, Michael Robern, Zahi Touma, Iyabode Tiamiyu, Oksana Gurtovaya, Alena Pechonkina, Afsaneh Mozaffarian, Bryan Downie, Franziska Matzkies, Daniel Wallace, Victoria P Werth, Roy Fleischmann, Michael Robern, Zahi Touma, Iyabode Tiamiyu, Oksana Gurtovaya, Alena Pechonkina, Afsaneh Mozaffarian, Bryan Downie, Franziska Matzkies, Daniel Wallace

Abstract

Objectives: To explore the safety and efficacy of filgotinib (FIL), a Janus kinase 1 inhibitor, and lanraplenib (LANRA), a spleen kinase inhibitor, in cutaneous lupus erythematosus (CLE).

Methods: This was a phase 2, randomized, double-blind, placebo-controlled, exploratory, proof-of-concept study of LANRA (30 mg), FIL (200 mg) or placebo (PBO) once daily for 12 weeks in patients with active CLE. At week 12, PBO patients were rerandomized 1:1 to receive LANRA or FIL for up to 36 additional weeks.

Results: Of 47 randomized patients, 45 were treated (PBO, n = 9; LANRA, n = 19; FIL, n = 17). The primary endpoint [change from baseline in Cutaneous Lupus Erythematosus Disease Area and Severity Index Activity (CLASI-A) score at week 12] was not met. The least squares mean CLASI-A score change from baseline was -5.5 (s.e. 2.56) with PBO, -4.5 (1.91) with LANRA and -8.7 (1.85) with FIL. Numerical differences between FIL and PBO were greater in select subgroups. A ≥5-point improvement in the CLASI-A score at week 12 was achieved by 50.0%, 56.3% and 68.8% in the PBO, LANRA and FIL arms, respectively. A numerically greater proportion of patients in the FIL arm (50%) also achieved ≥50% improvement in the CLASI-A score at week 12 (37.5% PBO, 31.3% LANRA). Most adverse events (AEs) were mild or moderate in severity. Two serious AEs were reported with LANRA and one with FIL.

Conclusion: The primary endpoint was not met. Select subgroups displayed a numerically greater treatment response to FIL relative to PBO. LANRA and FIL were generally well tolerated.

Trial registration: ClinicalTrials.gov identifier NCT03134222.

Keywords: clinical trials and methods; cytokines and inflammatory mediators; inflammation; skin; systemic lupus erythematosus and autoimmunity.

© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Rheumatology.

Figures

Fig . 1
Fig. 1
CLASI-A score least squares mean change from baseline to week 12 Full analysis set includes patients who were randomized and received at least one dose of study drug. Baseline value was the last available value collected on or prior to the day of the first dose of study drug. The adjusted means were obtained from a mixed effects model for repeated measures with baseline CLASI-A score, stratification factors, visit and treatment*visit as fixed effects and patient as a random effect.
Fig . 2
Fig. 2
Secondary and exploratory endpoints (change from baseline or meeting criteria at week 12) (A) Secondary endpoints: proportion of patients with a ≥5-point improvement in the CLASI-A score (left) and proportion with no worsening in the CLASI-A score (right). (B) Exploratory endpoints: percentage of patients with ≥50% improvement in the CLASI-A score and patient- and physician-reported QoL assessments. Full analysis set includes patients who were randomized and received at least one dose of study drug. Increase in TSQM indicates improvement. DLQI: Dermatology Life Quality Index; TSQM: Treatment Satisfaction Questionnaire for Medication; VAS: Visual Analog Scale.
Fig . 3
Fig. 3
Change in the CLASI-A score from baseline to week 12 by subgroup (A) Mean (95% CI) change from baseline for PBO, LANRA and FIL groups for selected subgroups. (B) Forest plot showing the median (Q1, Q3) for the PBO, LANRA and FIL groups for all subgroups. Age (in years) was calculated from date of first study drug administration. Disease subtype as reported in the clinical database was used for subgroup derivation. csDMARD subgroups determined per systemic DMARD use as reported in the clinical database. The following routes of administration were considered for systemic corticosteroids: oral, intravenous, intramuscular and subcutaneous. The duration of CLE was calculated based on the reported date of CLE diagnosis and the date of enrolment in the study. For all treatment groups, the baseline value was the last available value on or prior to the first dose of the study drug. *IFN activity was not a prespecified subgroup analysis. IFN activity was classified into subgroups according to whether the value was below (low) or above (high) the median value at baseline.

References

    1. Chen KL, Krain RL, Werth VP. Advancing understanding, diagnosis, and therapies for cutaneous lupus erythematosus within the broader context of systemic lupus erythematosus. F1000Res 2019;8:F1000 Faculty Rev-332.
    1. Rönnblom L, Leonard D. Interferon pathway in SLE: one key to unlocking the mystery of the disease. Lupus Sci Med 2019;6:e000270.
    1. Kuhn A, Bijl M. Pathogenesis of cutaneous lupus erythematosus. Lupus 2008;17:389–93.
    1. Little AJ, Vesely MD. Cutaneous lupus erythematosus: current and future pathogenesis-directed therapies. Yale J Biol Med 2020;93:81–95.
    1. Borucki R, Werth VP. Expert perspective: an evidence-based approach to refractory cutaneous lupus erythematosus. Arthritis Rheum 2020;72:1777–85.
    1. Furie R, Khamashta M, Merrill JT et al. Anifrolumab, an anti-interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheum 2017;69:376–86.
    1. Furie RA, Morand EF, Bruce IN et al. Type I interferon inhibitor anifrolumab in active systemic lupus erythematosus (TULIP-1): a randomised, controlled, phase 3 trial. Lancet Rheum 2019;1:e208–19.
    1. Morand EF, Furie R, Tanaka Y et al. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med 2020;382:211–21.
    1. Furie R, Werth VP, Merola JF et al. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus erythematosus. J Clin Invest 2019;129:1359–71.
    1. Werth V, Furie R, Romero-Diaz J et al. BIIB059, a humanized monoclonal antibody targeting BDCA2 on plasmacytoid dendritic cells (PDC), shows dose-related efficacy in the phase 2 LILAC study in patients (PTS) with active cutaneous lupus erythematosus (CLE). Ann Rheum Dis 2020;79:120–1.
    1. Alunno A, Padjen I, Fanouriakis A, Boumpas DT. Pathogenic and therapeutic relevance of JAK/STAT signaling in systemic lupus erythematosus: integration of distinct inflammatory pathways and the prospect of their inhibition with an oral agent. Cells 2019;8:898.
    1. Dey-Rao R, Smith JR, Chow S, Sinha AA. Differential gene expression analysis in CCLE lesions provides new insights regarding the genetics basis of skin vs. systemic disease. Genomics 2014;104:144–55.
    1. Taylor PC, Downie B, Elboudwarej E et al. Whole blood transcriptional changes following selective inhibition of Janus kinase 1 (JAK1) by filgotinib in adults with moderately-to-severely active rheumatoid arthritis with prior inadequate response to methotrexate (FINCH1). Ann Rheum Dis 2020;79:996–7.
    1. Taylor PC, Downie B, Elboudwarej E et al. Whole blood transcriptional changes following selective inhibition of Janus kinase 1 (JAK1) by filgotinib In MTX-naive adults with moderately-to-severely active rheumatoid arthritis (RA) (FINCH3). Ann Rheum Dis 2020;79:1017–8.
    1. Mócsai A, Ruland J, Tybulewicz VL. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol 2010;10:387–402.
    1. Braegelmann C, Hölzel M, Ludbrook V et al. Spleen tyrosine kinase (SYK) is a potential target for the treatment of cutaneous lupus erythematosus patients. Exp Dermatol 2016;25:375–9.
    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 Lett 2020;11:506–13.
    1. Askanase A, Byron M, Keyes-Elstein LL et al. Treatment of lupus nephritis with abatacept: the abatacept and cyclophosphamide combination efficacy and safety study. Arthritis Rheum 2014;66:3096–104.
    1. Furie R, Nicholls K, Cheng TT et al. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol 2014;66:379–89.
    1. Isenberg D, Furie R, Jones N et al. Efficacy, safety, and pharmacodynamic effects of the Bruton’s tyrosine kinase inhibitor, fenebrutinib (GDC-0853), in moderate to severe systemic lupus erythematosus: results of a phase 2 randomized controlled trial. Arthritis Rheumatol 2019;71(Suppl 10):abstract L15.
    1. Rovin BH, Furie R, Latinis K et al. Efficacy and safety of rituximab in patients with active proliferative lupus nephritis: the lupus nephritis assessment with rituximab study. Arthritis Rheum 2012;64:1215–26.
    1. Wallace DJ, Dörner T, Pisetsky D et al. Efficacy and safety of evobrutinib (M2951) in adult patients with systemic lupus erythematosus who received standard of care therapy: a phase ii, randomized, double-blind, placebo-controlled dose ranging study. Arthritis Rheumatol 2020;72(Suppl 10):abstract 0865.
    1. Merrill JT, Manzi S, Aranow C et al. Lupus community panel proposals for optimising clinical trials: 2018. Lupus Sci Med 2018;5:e000258.
    1. Chang AY, Ghazi E, Okawa J, Werth VP. Quality of life differences between responders and nonresponders in the treatment of cutaneous lupus erythematosus. JAMA Dermatol 2013;149:104–6.
    1. Klein R, Moghadam-Kia S, LoMonico J et al. Development of the CLASI as a tool to measure disease severity and responsiveness to therapy in cutaneous lupus erythematosus. Arch Dermatol 2011;147:203–8.
    1. Chakka S, Krain RL, Ahmed S et al. Evaluating change in disease activity needed to reflect meaningful improvement in quality of life for clinical trials in cutaneous lupus erythematosus. J Am Acad Dermatol 2021;84:1562–7.
    1. Werth V, Karnell JL, Rees W et al. Targeting plasmacytoid dendritic cells improves cutaneous lupus erythematosus skin lesions and reduces type I interferon levels: results of a phase 1 study of VIB7734. Arthritis Rheumatol 2020;72(Suppl 10):abstract L10.
    1. Merrill JW, Worth V, Furie R et al. Efficacy and safety of iberdomide in patients with active systemic lupus erythematosus: 24-week results of a phase 2, randomized, placebo-controlled study. Arthritis Rheumatol 2020;72(Suppl 10):abstract 0987.
    1. Furie R, van Vollenhoven R, Kalunian K et al. Efficacy and safety results from a phase 2, randomized, double-blind trial of BIIB059, an anti-BDCA2 antibody, in SLE. Arthritis Rheumatol 2020;72(Suppl 10):abstract 0935.
    1. Werth V, Furie R, Morand E et al. Early and sustained reduction in severity of skin disease with anifrolumab treatment in patients with active SLE measured by the Cutaneous Lupus Erythematosus Disease Area and Severity Index (CLASI): pooled data from 2 phase 3 studies. Arthritis Rheumatol 2020;72(Suppl 10):abstract 0985.
    1. Albrecht J, Taylor L, Berlin JA et al. The CLASI (Cutaneous Lupus Erythematosus Disease Area and Severity Index): an outcome instrument for cutaneous lupus erythematosus. J Invest Dermatol 2005;125:889–94.
    1. van Vollenhoven RF, Petri MA, Cervera R et al. Belimumab in the treatment of systemic lupus erythematosus: high disease activity predictors of response. Ann Rheum Dis 2012;71:1343–9.
    1. Coricello A, Mesiti F, Lupia A, Maruca A, Alcaro S. Inside perspective of the synthetic and computational toolbox of JAK inhibitors: recent updates. Molecules 2020;25: 3321.
    1. Harrington R, Al Nokhatha SA, Conway R. JAK inhibitors in rheumatoid arthritis: an evidence-based review on the emerging clinical data. J Inflamm Res 2020;13:519–31.
    1. Mease P, Coates LC, Helliwell PS et al. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active psoriatic arthritis (EQUATOR): results from a randomised, placebo-controlled, phase 2 trial. Lancet 2018;392:2367–77.
    1. Szilveszter KP, Németh T, Mócsai A. Tyrosine kinases in autoimmune and inflammatory skin diseases. Front Immunol 2019;10:1862.
    1. van der Heijde D, Baraliakos X, Gensler LS et al. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active ankylosing spondylitis (TORTUGA): results from a randomised, placebo-controlled, phase 2 trial. Lancet 2018;392:2378–87.
    1. Vermeire S, Schreiber S, Petryka R et al. Clinical remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet 2017;389:266–75.
    1. Xeljanz (tofacitinib) prescribing information. (11 December, date last accessed).
    1. Wallace DJ, Furie RA, Tanaka Y et al. Baricitinib for systemic lupus erythematosus: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2018;392:222–31.
    1. Pamuk ON, Tsokos GC. Spleen tyrosine kinase inhibition in the treatment of autoimmune, allergic and autoinflammatory diseases. Arthritis Res Ther 2010;12:222.
    1. Uzé G, Schreiber G, Piehler J, Pellegrini S. The receptor of the type I interferon family. Curr Top Microbiol Immunol 2007;316:71–95.
    1. Sarkar MK, Hile GA, Tsoi LC et al. Photosensitivity and type I IFN responses in cutaneous lupus are driven by epidermal-derived interferon kappa. Ann Rheum Dis 2018;77:1653–64.
    1. Tsoi LC, Hile GA, Berthier CC et al. Hypersensitive IFN responses in lupus keratinocytes reveal key mechanistic determinants in cutaneous lupus. J Immunol 2019;202:2121–30.
    1. Hinojosa-Azaola A, Romero-Diaz J, Vargas-Ruiz AG et al. Venous and arterial thrombotic events in systemic lupus erythematosus. J Rheum 2016;43:576–86.
    1. Tavilisse (fostamatinib disodium hexahydrate) prescribing information. (11 December, date last accessed).
    1. Genovese MC, Kalunian K, Gottenberg JE et al. Effect of filgotinib vs placebo on clinical response in patients with moderate to severe rheumatoid arthritis refractory to disease-modifying antirheumatic drug therapy: the FINCH 2 randomized clinical trial. JAMA 2019;322:315–25.

Source: PubMed

3
Abonnere