Relationship of anifrolumab pharmacokinetics with efficacy and safety in patients with systemic lupus erythematosus

Yen Lin Chia, Jianchun Zhang, Raj Tummala, Tomas Rouse, Richard A Furie, Eric F Morand, Yen Lin Chia, Jianchun Zhang, Raj Tummala, Tomas Rouse, Richard A Furie, Eric F Morand

Abstract

Objectives: To characterize the relationship of anifrolumab pharmacokinetics with efficacy and safety in patients with moderate to severe SLE despite standard therapy, using pooled data from two phase 3 trials.

Methods: TULIP-1 and TULIP-2 were randomized, placebo-controlled, 52-week trials of intravenous anifrolumab (every 4 weeks for 48 weeks). For the exposure-response analysis, BILAG-based Composite Lupus Assessment (BICLA) or SLE Responder Index [SRI(4)] response rates at week 52 in each quartile/tertile of average anifrolumab serum concentration (Cave) were compared for anifrolumab and placebo in all-comers, patients who completed treatment, and IFN gene signature (IFNGS)-high patients who completed treatment, using average marginal effect logistic regression. Relationships between exposure and key safety events were assessed graphically.

Results: Of patients in TULIP-1/TULIP-2 who received anifrolumab (150 mg, n = 91; 300 mg, n = 356) or placebo (n = 366), 574 completed treatment, of whom 470 were IFNGS high. In the exposure-efficacy analyses, BICLA and SRI(4) treatment differences favouring anifrolumab 300 mg vs placebo were observed across Cave subgroups and all analysis populations. Logistic regression identified Cave as a significant covariate for predicted BICLA response, as higher anifrolumab Cave predicted greater efficacy. There was no evidence of exposure-driven incidence of key safety events through week 52 in patients receiving anifrolumab 150 or 300 mg.

Conclusion: While higher Cave predicted greater efficacy, consistent positive benefit favouring anifrolumab 300 mg vs placebo was observed in BICLA and SRI(4) responses across Cave subgroups in the TULIP trials. There was no evidence of exposure-driven safety events.

Clinicaltrial.gov numbers: NCT02446912, NCT02446899.

Keywords: anifrolumab; autoimmunity; biologic therapies; clearance; efficacy; exposure–response; population pharmacokinetics; safety; systematic lupus erythematosus.

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

Figures

Fig . 1
Fig. 1
BICLA response at week 52 by analysis of populations in TULIP-1 and TULIP-2 Data were from the pooled exposure–response analysis set. Response rates and treatment difference for BICLA were calculated using the AME approach based on logistic regression models by treating quartile/median groups along with placebo group as one covariate, and stratification factors by SLEDAI-2K score at screening (vs ≥10 points), day 1 glucocorticoid dose (<10 mg/day vs ≥10 mg/day prednisone or equivalent), and type I IFNGS at screening (high vs low), whenever applicable. Tertiles (µg/ml) were defined as: G1 <31.2, G2 ≥31.2 to <43.8 and G3 ≥43.8; quartiles (µg/ml) were defined as: Q1 <27.6, Q2 ≥27.6 to <39.2, Q3 ≥39.2 to <49.8 and Q4 ≥49.8. AME: average marginal effect; BICLA: BILAG-based Composite Lupus Assessment; G: tertile; IFNGS: type I IFN gene signature; n: number of patients; N: number of patients in group; Q: quartile; SLEDAI-2K: SLE Disease Activity Index 2000.
Fig . 2
Fig. 2
Predicted BICLA response at week 52 in all patients and IFNGS-high patients who completed treatment Data were from the pooled exposure–response analysis set. Two hundred and twenty-seven IFNGS-high patients receiving placebo were included in the model but are not shown; Cave was set to 0 μg/ml. (A) All patients; (B) IFNGS-high patients. BICLA: BILAG–based Composite Lupus Assessment; Cave: average serum concentration; IFNGS: type I IFN gene signature; n: number of patients; Obs: observed; SLEDAI-2K: SLE Disease Activity Index 2000.
Fig . 3
Fig. 3
SRI(4) response at week 52 by analysis populations in TULIP-1 and TULIP-2 Data were from the pooled exposure–response analysis set. Response rates and treatment difference for SRI(4) were calculated using the AME approach based on logistic regression models by treating quartile/median groups along with placebo group as one covariate, and stratification factors by SLEDAI-2K score at screening (vs ≥10 points), day 1 oral glucocorticoid dose (<10 mg/day vs ≥10 mg/day prednisone or equivalent) and type I IFNGS at screening (high vs low), whenever applicable. Tertiles (µg/ml) were defined as: G1 <31.2, G2 ≥31.2 to <43.8 and G3 ≥43.8; quartiles (µg/ml) were defined as: Q1 <27.6, Q2 ≥27.6 to <39.2, Q3 ≥39.2 to <49.8 and Q4 ≥49.8. AME: average marginal effect; G: tertile; IFNGS: type I IFN gene signature; n: number of patients; N: number of patients in group; Q: quartile; SLEDAI-2K: SLE Disease Activity Index 2000; SRI(4): SLE Responder Index of ≥4.
Fig . 4
Fig. 4
Predicted SRI(4) response at week 52 in all patients and IFNGS-high patients who completed treatment Data were from the pooled exposure–response analysis set. Two hundred and seventy-six placebo patients (A) and 227 IFNGS-high placebo patients (B) from pooled TULIP trials who completed the treatment were included in the model but are not shown; Cave was set to 0 μg/ml. Cave: average serum concentration; IFNGS: type I IFN gene signature; n: number of patients; Obs: observed; SLEDAI-2K: SLE Disease Activity Index 2000; SRI(4): SLE Responder Index ≥4.
Fig . 5
Fig. 5
Incidence of key adverse events by PK medians/quartiles in all-comers in TULIP-1 and TULIP-2 Data were from the pooled exposure–response analysis set. The overall incidence rate included the patients excluded from the exposure–response analysis set. (A) Herpes zoster; (B) non-opportunisitic serious infections; (C) infusion-related reactions; (D) hypersensitivity. Cave: average anifrolumab concentrations up to the first incidence of serious infection or end of treatment; Cmax: latest anifrolumab peak concentrations at the end of infusion prior to first incidence of infusion-related events or end of treatment; n: number of patients; PK: pharmacokinetic.

References

    1. Samotij D, Reich A.. Biologics in the treatment of lupus erythematosus: a critical literature review. Biomed Res Int 2019;2019:8142368.
    1. Peng L, Oganesyan V, Wu H, Dall'Acqua WF, Damschroder MM.. Molecular basis for antagonistic activity of anifrolumab, an anti-interferon-α receptor 1 antibody. mAbs 2015;7:428–39.
    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.
    1. Petri M, Singh S, Tesfasyone H. et al. Longitudinal expression of type I interferon responsive genes in systemic lupus erythematosus. Lupus 2009;18:980–9.
    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.
    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 USA 2003;100:2610–5.
    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.
    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.
    1. Morand EF, Furie R, Tanaka Y. et al.; TULIP-2 Trial Investigators. Trial of anifrolumab in active systemic lupus erythematosus. N Engl J Med 2020;382:211–21.
    1. Furie R, Khamashta M, Merrill JT. et al.; CD1013 Study Investigators. Anifrolumab, an anti-interferon-α receptor monoclonal antibody, in moderate-to-severe systemic lupus erythematosus. Arthritis Rheumatol 2017;69:376–86.
    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.
    1. Toomula N, Kumar S, Kumar A, Phaneendra M.. Role of pharmacokinetic studies in drug discovery. J Bioequiv Availab 2011;3:263–7.
    1. Food and Drug Administration (FDA). Guidance for industry: Exposure-response relationships — study design, data analysis, and regulatory applications. Rockville, MD: FDA, 2003.
    1. Chia YL, Santiago L, Wang B. et al. Exposure–response analysis for selection of optimal dosage regimen of anifrolumab in patients with systemic lupus erythematosus. Rheumatology (Oxford) 2021; keab176.
    1. Kuruvilla D, Mai T, Tummala R, Roskos L, Chia YL. Characterization of the nonlinear pharmacokinetics and time-varying clearance of anifrolumab in patients with active systemic lupus erythematosus. American Association of Pharmaceutical Scientists (AAPS) PharmSci 360 Virtual Event 2020.. (September 2021, last accessed).
    1. Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum 1997;40:1725.
    1. Yao Y, Higgs BW, Richman L, White B, Jallal B.. 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.
    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.
    1. Tummala R, Abreu G, Pineda L, Michaels MA. 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.
    1. Morand EF, Furie R, Tanaka Y. et al. OP0049 Efficacy of anifrolumab in active systemic lupus erythematosus: patient subgroup analysis of BICLA response in 2 phase 3 trials. Ann Rheum Dis 2020;79(Suppl 1):32.
    1. Chia YL, Tummala R, Mai T. et al. Characterization of PK/PD of anifrolumab in patients with moderate to severe SLE. Ann Rheum Dis 2021;80(Suppl 1):590–1.
    1. Furie R, Kalunian K, Merrill J, Abreu G, Tummala R. Lupus disease activity after cessation of anifrolumab treatment during the phase 2b MUSE trial follow-up period. Arthritis Rheumatol 2020; 72(suppl 10): .
    1. Chatham WW, Furie R, Saxena A. et al. Long-term safety and efficacy of anifrolumab in adults with systemic lupus erythematosus: results of a phase II open-label extension study. Arthritis Rheumatol 2021;73:816–25.

Source: PubMed

3
Abonnieren