Anifrolumab efficacy and safety by type I interferon gene signature and clinical subgroups in patients with SLE: post hoc analysis of pooled data from two phase III trials

Edward M Vital, Joan T Merrill, Eric F Morand, Richard A Furie, Ian N Bruce, Yoshiya Tanaka, Susan Manzi, Kenneth C Kalunian, Rubana N Kalyani, Katie Streicher, Gabriel Abreu, Raj Tummala, Edward M Vital, Joan T Merrill, Eric F Morand, Richard A Furie, Ian N Bruce, Yoshiya Tanaka, Susan Manzi, Kenneth C Kalunian, Rubana N Kalyani, Katie Streicher, Gabriel Abreu, Raj Tummala

Abstract

Objectives: To characterise the efficacy and safety of anifrolumab in patients with systemic lupus erythematosus (SLE) according to interferon gene signature (IFNGS), demographic and clinical subgroups.

Methods: We performed post hoc analyses of pooled data from the 52-week phase III TULIP-1/TULIP-2 placebo-controlled trials of intravenous anifrolumab in moderate-to-severe SLE. Outcomes were assessed in predefined subgroups: IFNGS (high/low), age, sex, body mass index, race, geographic region, age of onset, glucocorticoid use, disease activity and serological markers.

Results: In pooled data, patients received anifrolumab 300 mg (360/726) or placebo (366/726); 82.6% were IFNGS-high. IFNGS-high patients had greater baseline disease activity and were more likely to have abnormal serological markers versus IFNGS-low patients. In the total population, a greater proportion of patients treated with anifrolumab versus placebo achieved British Isles Lupus Assessment Group-based Composite Lupus Assessment (BICLA) response at week 52 (difference 16.6%; nominal p<0.001). BICLA response treatment differences with anifrolumab versus placebo were comparable to the total population across most predefined subgroups, including subgroups for baseline glucocorticoid dosage (<10/≥10 mg/day prednisone/equivalent) and for clinical disease activity (SLE Disease Activity Index 2000 score <10/≥10). Subgroups with larger treatment differences included IFNGS-high patients (18.2%), patients with abnormal baseline serological markers (23.1%) and Asian patients (29.2%). The safety profile of anifrolumab was similar across subgroups.

Conclusions: Overall, this study supports the consistent efficacy and safety of anifrolumab across a range of patients with moderate-to-severe SLE. In a few subgroups, small sample sizes limited conclusions from being drawn regarding the treatment benefit with anifrolumab.

Trial registration number: NCT02446912, NCT02446899.

Keywords: biological therapy; lupus erythematosus; systemic; therapeutics.

Conflict of interest statement

Competing interests: EMV has received grant support from AstraZeneca, Roche/Genentech and Sandoz; received consulting fees from AstraZeneca, GlaxoSmithKline, Roche/Genentech, Aurinia and Sandoz; and was a speaker at a speaker bureau for Becton Dickinson and GlaxoSmithKline. JTM has received grant/research support from Bristol Myers Squibb and GlaxoSmithKline, and consultancy fees from AbbVie, Amgen, AstraZeneca, Aurinia, Bristol Myers Squibb, EMD Serono, GlaxoSmithKline, Janssen, Provention, Remegen and UCB. EFM has received grant support from, was a consultant for and was a speaker at a speaker bureau for AstraZeneca; received grant support and consulting fees from AbbVie, Bristol Myers Squibb, Eli Lilly, GlaxoSmithKline, Janssen, Merck Serono and UCB; received grant support from Bristol Myers Squibb and received consulting fees from Amgen, Biogen, CSL, Neovacs and Wolf Biotherapeutics. RAF has received grant/research support and consulting fees from AstraZeneca. INB is a National Institute for Health Research (NIHR) Senior Investigator Emeritus and is funded by the NIHR Manchester Biomedical Research Centre; has received grant/research support from Genzyme/Sanofi, GlaxoSmithKline, Roche and UCB; received consulting fees from Eli Lilly, GlaxoSmithKline, ILTOO, Merck Serono and UCB and was a speaker for AstraZeneca, GlaxoSmithKline and UCB. YT has received speaking fees and/or honoraria from AbbVie, Amgen, Astellas, AstraZeneca, Boehringer-Ingelheim, Bristol Myers Squibb, Chugai, Eisai, Eli Lilly, Gilead, Mitsubishi-Tanabe and YL Biologics, and has received research grants from AbbVie, Asahi-Kasei, Chugai, Boehringer-Ingelheim, Corrona, Daiichi-Sankyo, Eisai, Kowa, Mitsubishi-Tanabe and Takeda. SM has received grant/research support and consulting fees from AstraZeneca. KCK has received consulting fees from AstraZeneca. RNK, KS, GA and RJ are employees of AstraZeneca.

© Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
BICLA response at week 52 by subgroup. aSerological markers refer to anti-dsDNA antibodies (positive or negative), C3 (low or normal) and C4 (low or normal). The percentage of responders, the difference in estimates, associated 95% CIs and nominal p values were calculated using a stratified Cochran-Mantel-Haenszel approach, with stratification factors of SLEDAI-2K score at screening, baseline GC dosage, type I IFNGS test result at screening and study. Anti-dsDNA, antidouble-stranded DNA; BICLA, British Isles Lupus Assessment Group-based Composite Lupus Assessment; BMI, body mass index; C, complement; CI, confidence interval; GC, oral glucocorticoid; IFNGS, interferon gene signature; n, number of responders; N, number of patients in group; SLEDAI-2K, Systemic Lupus Erythematosus Disease Activity Index 2000.
Figure 2
Figure 2
Adjusted difference in cumulative percentages of patients with ≥1 AE during treatment with anifrolumab 300 mg versus placebo by subgroup in pooled TULIP-1 and TULIP-2 data. aSerological markers refer to anti-dsDNA antibodies (positive or negative), C3 (low or normal) and C4 (low or normal). Percentages indicate cumulative proportions that were adjusted using the Cochran-Mantel-Haenszel approach. Anti-dsDNA, antidouble-stranded DNA; AE, adverse event; BMI, body mass index; C, complement; CI, confidence interval; GC, oral glucocorticoid; IFNGS, interferon gene signature; n, number of responders; N, number of patients in group; SLEDAI-2K, Systemic Lupus Erythematosus Disease Activity Index 2000.
Figure 3
Figure 3
Adjusted difference in cumulative percentages of patients with ≥1 SAE during treatment with anifrolumab 300 mg versus placebo by subgroup in pooled TULIP-1 and TULIP-2 data. aSerological markers refer to anti-dsDNA antibodies (positive or negative), C3 (low or normal) and C4 (low or normal). Percentages indicate cumulative proportions that were adjusted using the Cochran-Mantel-Haenszel approach. Anti-dsDNA, anti-double-stranded DNA; BMI, body mass index; C, complement; CI, confidence interval; GC, oral glucocorticoid; IFNGS, interferon gene signature; n, number of responders; N, number of patients in group; SAE, serious adverse event; SLEDAI-2K, Systemic Lupus Erythematosus Disease Activity Index 2000.
Figure 4
Figure 4
BICLA response estimates and SEs from weeks 4–52 in (A) type I IFNGS-high and (B) IFNGS-low patients in pooled TULIP data. The percentage of responders was calculated using a stratified Cochran-Mantel-Haenszel approach, with stratification factors SLEDAI-2K score at screening, baseline GC dosage and study. Points represent response estimates plotted with SE. *Nominal p

Figure 5

Sustained oral GC taper by…

Figure 5

Sustained oral GC taper by subgroup for patients with SLE in pooled data…

Figure 5
Sustained oral GC taper by subgroup for patients with SLE in pooled data from the TULIP-1 and TULIP-2 trials. A sustained oral GC taper was defined as a dosage reduction to ≤7.5 mg/day from week 40 to week 52 in patients receiving ≥10 mg/day at baseline (prednisone or equivalent). The percentage of responders, the difference in estimates, associated 95% CIs and nominal p values were calculated using a stratified Cochran-Mantel-Haenszel approach, with stratification factors of SLEDAI-2K score at screening, type I IFNGS test result at screening and study. Anti-dsDNA, antidouble-stranded DNA; BMI, body mass index; C, complement; CI, confidence interval; GC, glucocorticoid; IFNGS, interferon gene signature; SLE, systemic lupus erythematosus; SLEDAI-2K, SLE Disease Activity Index 2000.

Figure 6

Annualised flare rate through week…

Figure 6

Annualised flare rate through week 52 by subgroup for patients with SLE in…

Figure 6
Annualised flare rate through week 52 by subgroup for patients with SLE in pooled data from the TULIP-1 and TULIP-2 trials. A flare is defined as either ≥1 new BILAG-2004 A or ≥2 new BILAG-2004 B items compared with the previous visit. The annualised flare rate was calculated using a negative binomial regression model, which included covariates of treatment group, stratification factors and study, and was adjusted for variations in exposure time. Anti-dsDNA, antidouble-stranded DNA; BILAG-2004, British Isles Lupus Assessment Group-2004; BMI, body mass index; C, complement; CI, confidence interval; GC, glucocorticoid; IFNGS, interferon gene signature; SLE, systemic lupus erythematosus; SLEDAI-2K, SLE Disease Activity Index 2000.
Figure 5
Figure 5
Sustained oral GC taper by subgroup for patients with SLE in pooled data from the TULIP-1 and TULIP-2 trials. A sustained oral GC taper was defined as a dosage reduction to ≤7.5 mg/day from week 40 to week 52 in patients receiving ≥10 mg/day at baseline (prednisone or equivalent). The percentage of responders, the difference in estimates, associated 95% CIs and nominal p values were calculated using a stratified Cochran-Mantel-Haenszel approach, with stratification factors of SLEDAI-2K score at screening, type I IFNGS test result at screening and study. Anti-dsDNA, antidouble-stranded DNA; BMI, body mass index; C, complement; CI, confidence interval; GC, glucocorticoid; IFNGS, interferon gene signature; SLE, systemic lupus erythematosus; SLEDAI-2K, SLE Disease Activity Index 2000.
Figure 6
Figure 6
Annualised flare rate through week 52 by subgroup for patients with SLE in pooled data from the TULIP-1 and TULIP-2 trials. A flare is defined as either ≥1 new BILAG-2004 A or ≥2 new BILAG-2004 B items compared with the previous visit. The annualised flare rate was calculated using a negative binomial regression model, which included covariates of treatment group, stratification factors and study, and was adjusted for variations in exposure time. Anti-dsDNA, antidouble-stranded DNA; BILAG-2004, British Isles Lupus Assessment Group-2004; BMI, body mass index; C, complement; CI, confidence interval; GC, glucocorticoid; IFNGS, interferon gene signature; SLE, systemic lupus erythematosus; SLEDAI-2K, SLE Disease Activity Index 2000.

References

    1. Dörner T, Furie R. Novel paradigms in systemic lupus erythematosus. Lancet 2019;393:2344–58. 10.1016/S0140-6736(19)30546-X
    1. Tesser A, de Carvalho LM, Sandrin-Garcia P, et al. . Higher interferon score and normal complement levels may identify a distinct clinical subset in children with systemic lupus erythematosus. Arthritis Res Ther 2020;22:91. 10.1186/s13075-020-02161-8
    1. Tirosh I, Spielman S, Barel O, et al. . Whole exome sequencing in childhood-onset lupus frequently detects single gene etiologies. Pediatr Rheumatol Online J 2019;17:52. 10.1186/s12969-019-0349-y
    1. Mina R, Brunner HI. Pediatric lupus--are there differences in presentation, genetics, response to therapy, and damage accrual compared with adult lupus? Rheum Dis Clin North Am 2010;36:53–80. vii–viii. 10.1016/j.rdc.2009.12.012
    1. Lewis MJ, Jawad AS. The effect of ethnicity and genetic ancestry on the epidemiology, clinical features and outcome of systemic lupus erythematosus. Rheumatology 2017;56:i67–77. 10.1093/rheumatology/kew399
    1. Pons-Estel GJ, Alarcón GS, Scofield L, et al. . Understanding the epidemiology and progression of systemic lupus erythematosus. Semin Arthritis Rheum 2010;39:257–68. 10.1016/j.semarthrit.2008.10.007
    1. Ginzler E, Guedes Barbosa LS, D'Cruz D. EMBRACE: phase 3/4, randomized, 52-week study of belimumab efficacy and safety in patients of black African ancestry with systemic lupus erythematosus. Arthritis Rheumatol 2021.
    1. Bengtsson AA, Sturfelt G, Truedsson L, et al. . Activation of type I interferon system in systemic lupus erythematosus correlates with disease activity but not with antiretroviral antibodies. Lupus 2000;9:664–71. 10.1191/096120300674499064
    1. Kirou KA, Lee C, George S, et al. . Activation of the interferon-alpha pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum 2005;52:1491–503. 10.1002/art.21031
    1. Feng X, Wu H, Grossman JM, et al. . Association of increased interferon-inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum 2006;54:2951–62. 10.1002/art.22044
    1. Psarras A, Alase A, Antanaviciute A, et al. . Functionally impaired plasmacytoid dendritic cells and non-haematopoietic sources of type I interferon characterize human autoimmunity. Nat Commun 2020;11:6149. 10.1038/s41467-020-19918-z
    1. Rodero MP, Decalf J, Bondet V, et al. . Detection of interferon alpha protein reveals differential levels and cellular sources in disease. J Exp Med 2017;214:1547–55. 10.1084/jem.20161451
    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 Rheumatol 2017;69:376–86. 10.1002/art.39962
    1. Yao Y, Higgs BW, Richman L, et al. . Use of type I interferon-inducible mRNAs as pharmacodynamic markers and potential diagnostic markers in trials with sifalimumab, an anti-IFNα antibody, in systemic lupus erythematosus. Arthritis Res Ther 2010;12(Suppl 1):S6. 10.1186/ar2887
    1. Merrill JT, Immermann F, Whitley M, et al. . The biomarkers of lupus disease study: a bold approach may mitigate interference of background immunosuppressants in clinical trials. Arthritis Rheumatol 2017;69:1257–66. 10.1002/art.40086
    1. Higgs BW, Liu Z, White B, et al. . Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann Rheum Dis 2011;70:2029–36. 10.1136/ard.2011.150326
    1. Peng L, Oganesyan V, Wu H, et al. . Molecular basis for antagonistic activity of anifrolumab, an anti-interferon-α receptor 1 antibody. MAbs 2015;7:428–39. 10.1080/19420862.2015.1007810
    1. Riggs JM, Hanna RN, Rajan B, et al. . Characterisation of anifrolumab, a fully human anti-interferon receptor antagonist antibody for the treatment of systemic lupus erythematosus. Lupus Sci Med 2018;5:e000261. 10.1136/lupus-2018-000261
    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. 10.1056/NEJMoa1912196
    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 Rheumatol 2019;1:e208–19. 10.1016/S2665-9913(19)30076-1
    1. Tanaka Y, Tummala R. Anifrolumab, a monoclonal antibody to the type I interferon receptor subunit 1, for the treatment of systemic lupus erythematosus: an overview from clinical trials. Mod Rheumatol 2021;31:1–12. 10.1080/14397595.2020.1812201
    1. AstraZeneca Pharmaceuticals LP . 2021. SAPHNELO™ (anifrolumab-fnia) [Prescribing Information]. Wilmington, DE.
    1. AstraZeneca . Saphnelo approved in Japan for systemic lupus erythematosus [press release]. Available: [Accessed February 14, 2022].
    1. AstraZeneca . Saphnelo recommended for approval in the EU by CHMP for the treatment of patients with systemic lupus erythematosus [press release]. Available: [Accessed 14 Feb 2022].
    1. Wallace DJ, Kalunian K, Petri MA, et al. . Efficacy and safety of epratuzumab in patients with moderate/severe active systemic lupus erythematosus: results from EMBLEM, a phase IIb, randomised, double-blind, placebo-controlled, multicentre study. Ann Rheum Dis 2014;73:183–90. 10.1136/annrheumdis-2012-202760
    1. Furie RA, Petri MA, Wallace DJ, et al. . Novel evidence-based systemic lupus erythematosus responder index. Arthritis Rheum 2009;61:1143–51. 10.1002/art.24698
    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. 10.1111/j.0022-202X.2005.23889.x
    1. Kosinski M, Gajria K, Fernandes AW, et al. . Qualitative validation of the FACIT-fatigue scale in systemic lupus erythematosus. Lupus 2013;22:422–30. 10.1177/0961203313476360
    1. McHorney CA, Ware JE, Raczek AE. The MOS 36-Item short-form health survey (SF-36): II. psychometric and clinical tests of validity in measuring physical and mental health constructs. Med Care 1993;31:247–63. 10.1097/00005650-199303000-00006
    1. Stokes ME, Davis CS, Koch GG. Categorical data analysis using SAS®. 3rd edn. SAS Institute, 2012.
    1. Tummala R, Abreu G, Pineda L, et al. . Safety profile of anifrolumab in patients with active SLE: an integrated analysis of phase II and III trials. Lupus Sci Med 2021;8:e000464. 10.1136/lupus-2020-000464
    1. Baechler EC, Batliwalla FM, Karypis G, et al. . Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc Natl Acad Sci U S A 2003;100:2610–5. 10.1073/pnas.0337679100
    1. Oke V, Gunnarsson I, Dorschner J, et al. . High levels of circulating interferons type I, type II and type III associate with distinct clinical features of active systemic lupus erythematosus. Arthritis Res Ther 2019;21:107. 10.1186/s13075-019-1878-y
    1. El-Sherbiny YM, Md Yusof MY, Psarras A, et al. . B cell tetherin: a flow cytometric cell-specific assay for response to type I interferon predicts clinical features and flares in systemic lupus erythematosus. Arthritis Rheumatol 2020;72:769–79. 10.1002/art.41187
    1. El-Sherbiny YM, Psarras A, Md Yusof MY, et al. . A novel two-score system for interferon status segregates autoimmune diseases and correlates with clinical features. Sci Rep 2018;8:5793. 10.1038/s41598-018-24198-1
    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. 10.1136/annrheumdis-2018-213197
    1. Stannard JN, Reed TJ, Myers E, et al. . Lupus skin is primed for IL-6 inflammatory responses through a keratinocyte-mediated autocrine type I interferon loop. J Invest Dermatol 2017;137:115–22. 10.1016/j.jid.2016.09.008
    1. van Holten J, Smeets TJM, Blankert P, et al. . Expression of interferon beta in synovial tissue from patients with rheumatoid arthritis: comparison with patients with osteoarthritis and reactive arthritis. Ann Rheum Dis 2005;64:1780–2. 10.1136/ard.2005.040477
    1. Zayat AS, Mahmoud K, Md Yusof MY, et al. . Defining inflammatory musculoskeletal manifestations in systemic lupus erythematosus. Rheumatology 2019;58:304–12. 10.1093/rheumatology/key277
    1. Mahmoud K, Zayat AS, Yusof Y, et al. . Responsiveness of clinical and ultrasound outcome measures in musculoskeletal systemic lupus erythematosus. Rheumatology 2019;58:1353–60. 10.1093/rheumatology/key422
    1. Mahmoud K, Zayat AS, Yusof MYM, et al. . Ultrasound to identify systemic lupus erythematosus patients with mUsculoskeletal symptoms who respond best to therapy: the US evaluation for mUsculoskeletal lupus longitudinal multicentre study. Rheumatology 2021;60:5194–204. 10.1093/rheumatology/keab288
    1. Little J, Parker B, Lunt M, et al. . Glucocorticoid use and factors associated with variability in this use in the systemic lupus international collaborating clinics inception cohort. Rheumatology 2018;57:677–87. 10.1093/rheumatology/kex444
    1. Isenberg D, Appel GB, Contreras G, et al. . Influence of race/ethnicity on response to lupus nephritis treatment: the ALMS study. Rheumatology 2010;49:128–40. 10.1093/rheumatology/kep346
    1. Litwic AE, Sriranganathan MK, Edwards CJ. Race and the response to therapies for lupus: how strong is the evidence? Int J Clin Rheumtol 2013;8:471–81. 10.2217/ijr.13.41
    1. Weckerle CE, Franek BS, Kelly JA, et al. . Network analysis of associations between serum interferon-α activity, autoantibodies, and clinical features in systemic lupus erythematosus. Arthritis Rheum 2011;63:1044–53. 10.1002/art.30187
    1. Iwamoto T, Dorschner J, Jolly M, et al. . Associations between type I interferon and antiphospholipid antibody status differ between ancestral backgrounds. Lupus Sci Med 2018;5:e000246. 10.1136/lupus-2017-000246
    1. Ko K, Koldobskaya Y, Rosenzweig E, et al. . Activation of the interferon pathway is dependent upon autoantibodies in African-American SLE patients, but not in European-American SLE patients. Front Immunol 2013;4:309. 10.3389/fimmu.2013.00309
    1. Casey KA, Smith MA, Sinibaldi D, et al. . Modulation of cardiometabolic disease markers by type I interferon inhibition in systemic lupus erythematosus. Arthritis Rheumatol 2021;73:459–71. 10.1002/art.41518

Source: PubMed

Подписаться