Dual IL-17A and IL-17F neutralisation by bimekizumab in psoriatic arthritis: evidence from preclinical experiments and a randomised placebo-controlled clinical trial that IL-17F contributes to human chronic tissue inflammation

Sophie Glatt, Dominique Baeten, Terry Baker, Meryn Griffiths, Lucian Ionescu, Alastair D G Lawson, Ash Maroof, Ruth Oliver, Serghei Popa, Foteini Strimenopoulou, Pavan Vajjah, Mark I L Watling, Nataliya Yeremenko, Pierre Miossec, Stevan Shaw, Sophie Glatt, Dominique Baeten, Terry Baker, Meryn Griffiths, Lucian Ionescu, Alastair D G Lawson, Ash Maroof, Ruth Oliver, Serghei Popa, Foteini Strimenopoulou, Pavan Vajjah, Mark I L Watling, Nataliya Yeremenko, Pierre Miossec, Stevan Shaw

Abstract

Objective: Interleukin (IL)-17A has emerged as pivotal in driving tissue pathology in immune-mediated inflammatory diseases. The role of IL-17F, sharing 50% sequence homology and overlapping biological function, remains less clear. We hypothesised that IL-17F, together with IL-17A, contributes to chronic tissue inflammation, and that dual neutralisation may lead to more profound suppression of inflammation than inhibition of IL-17A alone.

Methods: Preclinical experiments assessed the role of IL-17A and IL-17F in tissue inflammation using disease-relevant human cells. A placebo-controlled proof-of-concept (PoC) clinical trial randomised patients with psoriatic arthritis (PsA) to bimekizumab (n=39) or placebo (n=14). Safety, pharmacokinetics and clinical efficacy of multiple doses (weeks 0, 3, 6 (240 mg/160 mg/160 mg; 80 mg/40 mg/40 mg; 160 mg/80 mg/80 mg and 560 mg/320 mg/320 mg)) of bimekizumab, a humanised monoclonal IgG1 antibody neutralising both IL-17A and IL-17F, were investigated.

Results: IL-17F induced qualitatively similar inflammatory responses to IL-17A in skin and joint cells. Neutralisation of IL-17A and IL-17F with bimekizumab more effectively suppressed in vitro cytokine responses and neutrophil chemotaxis than inhibition of IL-17A or IL-17F alone. The PoC trial met both prespecified efficacy success criteria and showed rapid, profound responses in both joint and skin (pooled top three doses vs placebo at week 8: American College of Rheumatology 20% response criteria 80.0% vs 16.7% (posterior probability >99%); Psoriasis Area and Severity Index 100% response criteria 86.7% vs 0%), sustained to week 20, without unexpected safety signals.

Conclusions: These data support IL-17F as a key driver of human chronic tissue inflammation and the rationale for dual neutralisation of IL-17A and IL-17F in PsA and related conditions.

Trial registration number: NCT02141763; Results.

Keywords: autoimmune diseases; cytokines; inflammation; psoriatic arthritis.

Conflict of interest statement

Competing interests: DB, TB, MG, SG, LI, ADGL, AM, RO, SS, FS, PV, MILW are employees of UCB Pharma. DB, TB, ADGL, PV hold stocks and/or stock options in UCB Pharma. DB is a part-time employee of UCB Pharma and holds a part-time position at the Academic Medical Center/University of Amsterdam. DB received a grant from UCB Pharma to conduct preclinical experiments; DB received grants and/or consultant or investigator fees from the following organizations outside of the submitted work: AbbVie, Pfizer, MSD, Roche, BMS, Novartis, Eli Lilly, Boehringer Ingelheim and Glenmark. MG is a paid contractor for UCB working in a consulting capacity. PM is a scientific advisor to UCB Pharma and received associated fees outside of the submitted work. SP, NY declare no relevant conflicts of interest.

© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.

Figures

Figure 1
Figure 1
IL-17F contributes to inflammation and bimekizumab demonstrates superior efficacy relative to inhibition of IL-17A or IL-17F alone. Recombinant IL-17A and IL-17F, with or without TNF, were used to activate either (A) PsA synoviocytes (n=2) or (B) NHDFs (n=4), and IL-8 release was assessed following overnight culture. To evaluate the individual and collective influence of IL-17A and IL-17F, (C) PsA synoviocytes (n=4), (D, F, G) primary NHDFs (n=4) or (E) normal synoviocytes (n=5) were stimulated with Th17 supernatant with or without IL-17-specific blocking antibodies. Following overnight culture, either (C, D) inhibition of IL-8 production, (E, F) gene transcriptional changes or (G) inhibition of chemotactic potential was evaluated. For transcriptional analysis (E, F), genes were normalised to GAPDH mRNA and expressed as relative fold changes compared with unstimulated cells. For primary NHDFs, the panel presents genes that had a fold change ≥3 for Th17 stimulation under IgG conditions. Values were subject to conditional formatting (three colour scale: maximum value—red, minimum value—green, median value—yellow) for each target gene across groups. The same genes were analysed for normal synoviocytes and the conditional formatting was applied for genes expressing a fold change ≥3. Data are mean ±SEM. Figures are representative of three independent experiments. *represents a significant reduction versus IgG control, *P

Figure 2

Pharmacokinetic parameters of bimekizumab (PK-PPS).…

Figure 2

Pharmacokinetic parameters of bimekizumab (PK-PPS). (A) GeoMean plasma concentration–time profile of bimekizumab (PK-PPS)…

Figure 2
Pharmacokinetic parameters of bimekizumab (PK-PPS). (A) GeoMean plasma concentration–time profile of bimekizumab (PK-PPS) and (B) table of pharmacokinetic variables of bimekizumab. PK-PPS, pharmacokinetic per-protocol set. Note: Means, SDs and CVs were only calculated if at least one-third of the concentrations were quantified at a respective time point. CV, coefficient of variation.

Figure 3

ACR and PASI response rates…

Figure 3

ACR and PASI response rates by treatment group (PD-PPS). (A–C) ACR response rates…

Figure 3
ACR and PASI response rates by treatment group (PD-PPS). (A–C) ACR response rates (%, with 95% CI) and (D-E) PASI response rates (%, with 95% CI). Dashed vertical lines indicate drug intake (weeks 0, 3 and 6). Note: top three doses of bimekizumab (160 mg/80 mg/80 mg, 240 mg/160 mg/160 mg, 560 mg/320 mg/320 mg); PASI75 and PASI100 were only calculated for patients with BSA ≥3% psoriasis involvement at baseline. Modifications in concomitant therapy were permitted post week 8. Subject 003–00320 was excluded from the analyses for all time points after week 8 due to receiving an increased dose of naproxen, adalimumab and leflunomide for control of worsening disease. ACR20, American College of Rheumatology 20% response criteria; ACR50, American College of Rheumatology 50% response criteria; ACR70, American College of Rheumatology 70% response criteria; PASI75, Psoriasis Area and Severity Index 75% response criteria; PASI100, Psoriasis Area and Severity Index 100% response criteria; PD-PPS, pharmacodynamic per-protocol set.
Figure 2
Figure 2
Pharmacokinetic parameters of bimekizumab (PK-PPS). (A) GeoMean plasma concentration–time profile of bimekizumab (PK-PPS) and (B) table of pharmacokinetic variables of bimekizumab. PK-PPS, pharmacokinetic per-protocol set. Note: Means, SDs and CVs were only calculated if at least one-third of the concentrations were quantified at a respective time point. CV, coefficient of variation.
Figure 3
Figure 3
ACR and PASI response rates by treatment group (PD-PPS). (A–C) ACR response rates (%, with 95% CI) and (D-E) PASI response rates (%, with 95% CI). Dashed vertical lines indicate drug intake (weeks 0, 3 and 6). Note: top three doses of bimekizumab (160 mg/80 mg/80 mg, 240 mg/160 mg/160 mg, 560 mg/320 mg/320 mg); PASI75 and PASI100 were only calculated for patients with BSA ≥3% psoriasis involvement at baseline. Modifications in concomitant therapy were permitted post week 8. Subject 003–00320 was excluded from the analyses for all time points after week 8 due to receiving an increased dose of naproxen, adalimumab and leflunomide for control of worsening disease. ACR20, American College of Rheumatology 20% response criteria; ACR50, American College of Rheumatology 50% response criteria; ACR70, American College of Rheumatology 70% response criteria; PASI75, Psoriasis Area and Severity Index 75% response criteria; PASI100, Psoriasis Area and Severity Index 100% response criteria; PD-PPS, pharmacodynamic per-protocol set.

References

    1. Doss GP, Agoramoorthy G, Chakraborty C. TNF/TNFR: drug target for autoimmune diseases and immune-mediated inflammatory diseases. Front Biosci 2014;19:1028–40. 10.2741/4265
    1. Zhao R, Zhou H, Su SB. A critical role for interleukin-1β in the progression of autoimmune diseases. Int Immunopharmacol 2013;17:658–69. 10.1016/j.intimp.2013.08.012
    1. Ho LJ, Luo SF, Lai JH. Biological effects of interleukin-6: clinical applications in autoimmune diseases and cancers. Biochem Pharmacol 2015;97:16–26. 10.1016/j.bcp.2015.06.009
    1. Durham LE, Kirkham BW, Taams LS. Contribution of the IL-17 pathway to psoriasis and psoriatic arthritis. Curr Rheumatol Rep 2015;17:55 10.1007/s11926-015-0529-9
    1. Raychaudhuri SP, Raychaudhuri SK. Mechanistic rationales for targeting interleukin-17A in spondyloarthritis. Arthritis Res Ther 2017;19:51 10.1186/s13075-017-1249-5
    1. Hymowitz SG, Filvaroff EH, Yin JP, et al. . IL-17s adopt a cystine knot fold: structure and activity of a novel cytokine, IL-17F, and implications for receptor binding. Embo J 2001;20:5332–41. 10.1093/emboj/20.19.5332
    1. Yang XO, Chang SH, Park H, et al. . Regulation of inflammatory responses by IL-17F. J Exp Med 2008;205:1063–75. 10.1084/jem.20071978
    1. Jin W, Dong C. IL-17 cytokines in immunity and inflammation. Emerg Microbes Infect 2013;2:e60 10.1038/emi.2013.58
    1. Wright JF, Bennett F, Li B, et al. . The human IL-17F/IL-17A heterodimeric cytokine signals through the IL-17RA/IL-17RC receptor complex. J Immunol 2008;181:2799–805. 10.4049/jimmunol.181.4.2799
    1. Ota K, Kawaguchi M, Matsukura S, et al. . Potential involvement of IL-17F in asthma. J Immunol Res 2014;2014:602846 10.1155/2014/602846
    1. Iboshi Y, Nakamura K, Fukaura K, et al. . Increased IL-17A/IL-17F expression ratio represents the key mucosal T helper/regulatory cell-related gene signature paralleling disease activity in ulcerative colitis. J Gastroenterol 2017;52:315–26. 10.1007/s00535-016-1221-1
    1. Iwakura Y, Ishigame H, Saijo S, et al. . Functional specialization of interleukin-17 family members. Immunity 2011;34:149–62. 10.1016/j.immuni.2011.02.012
    1. Wilson NJ, Boniface K, Chan JR, et al. . Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 2007;8:950–7. 10.1038/ni1497
    1. Hot A, Miossec P. Effects of interleukin (IL)-17A and IL-17F in human rheumatoid arthritis synoviocytes. Ann Rheum Dis 2011;70:727–32. 10.1136/ard.2010.143768
    1. Mease PJ, Fleischmann R, Deodhar AA, et al. . Effect of certolizumab pegol on signs and symptoms in patients with psoriatic arthritis: 24-week results of a Phase 3 double-blind randomised placebo-controlled study (RAPID-PsA). Ann Rheum Dis 2014;73:48–55. 10.1136/annrheumdis-2013-203696
    1. Johansen C, Usher PA, Kjellerup RB, et al. . Characterization of the interleukin-17 isoforms and receptors in lesional psoriatic skin. Br J Dermatol 2009;160:319–24. 10.1111/j.1365-2133.2008.08902.x
    1. Fujishima S, Watanabe H, Kawaguchi M, et al. . Involvement of IL-17F via the induction of IL-6 in psoriasis. Arch Dermatol Res 2010;302:499–505. 10.1007/s00403-010-1033-8
    1. van Baarsen LG, Lebre MC, van der Coelen D, et al. . Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and osteoarthritis: possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res Ther 2014;16:426 10.1186/s13075-014-0426-z
    1. Zrioual S, Toh ML, Tournadre A, et al. . IL-17RA and IL-17RC receptors are essential for IL-17A-induced ELR+ CXC chemokine expression in synoviocytes and are overexpressed in rheumatoid blood. J Immunol 2008;180:655–63. 10.4049/jimmunol.180.1.655
    1. Raychaudhuri SP, Raychaudhuri SK, Genovese MC. IL-17 receptor and its functional significance in psoriatic arthritis. Mol Cell Biochem 2012;359:419–29. 10.1007/s11010-011-1036-6
    1. Belasco J, Louie JS, Gulati N, et al. . Comparative genomic profiling of synovium versus skin lesions in psoriatic arthritis. Arthritis Rheumatol 2015;67:934–44. 10.1002/art.38995
    1. Kuo HL, Huang CC, Lin TY, et al. . IL-17 and CD40 ligand synergistically stimulate the chronicity of diabetic nephropathy. Nephrol Dial Transplant 2017. (Epub ahead of print: 1 Mar 2017). 10.1093/ndt/gfw397
    1. Teunissen MB, Koomen CW, de Waal Malefyt R, et al. . Interleukin-17 and interferon-gamma synergize in the enhancement of proinflammatory cytokine production by human keratinocytes. J Invest Dermatol 1998;111:645–9. 10.1046/j.1523-1747.1998.00347.x
    1. Kolbinger F, Loesche C, Valentin MA, et al. . β-Defensin 2 is a responsive biomarker of IL-17A-driven skin pathology in patients with psoriasis. J Allergy Clin Immunol 2017;139:923–32.e8 10.1016/j.jaci.2016.06.038
    1. Glatt S, Helmer E, Haier B, et al. . First-in-human randomized study of bimekizumab, a humanized monoclonal antibody and selective dual inhibitor of IL-17A and IL-17F, in mild psoriasis. Br J Clin Pharmacol 2017;83:991–1001. 10.1111/bcp.13185
    1. McInnes IB, Mease PJ, Kirkham B, et al. . Secukinumab, a human anti-interleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015;386:1137–46. 10.1016/S0140-6736(15)61134-5
    1. Targan SR, Feagan B, Vermeire S, et al. . A Randomized, double-blind, placebo-controlled phase 2 study of brodalumab in patients with moderate-to-severe crohn’s disease. Am J Gastroenterol 2016;111:1599–607. 10.1038/ajg.2016.298
    1. Mease PJ, van der Heijde D, Ritchlin CT, et al. . Ixekizumab, an interleukin-17A specific monoclonal antibody, for the treatment of biologic-naive patients with active psoriatic arthritis: results from the 24-week randomised, double-blind, placebo-controlled and active (adalimumab)-controlled period of the phase III trial SPIRIT-P1. Ann Rheum Dis 2017;76:79–87. 10.1136/annrheumdis-2016-209709
    1. Roman M, Madkan VK, Chiu MW. Profile of secukinumab in the treatment of psoriasis: current perspectives. Ther Clin Risk Manag 2015;11:1767–77. 10.2147/TCRM.S79053
    1. Mease PJ, Genovese MC, Greenwald MW, et al. . Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med 2014;370:2295–306. 10.1056/NEJMoa1315231

Source: PubMed

3
Abonnere