Out of the shadow of interleukin-17A: the role of interleukin-17F and other interleukin-17 family cytokines in spondyloarthritis

Nataliya Yeremenko, Nataliya Yeremenko

Abstract

Purpose of review: The last decade has witnessed tremendous advances in revealing an important role for the interleukin (IL)-17 cytokine family in the pathogenesis of spondyloarthritis (SpA). Although most attention has been focused on IL-17A, a potential role of other IL-17 family members in inflammation and tissue remodelling is emerging. Herein, I review recent studies covering the role of IL-17B-F cytokines in the pathogenesis of SpA.

Recent findings: Several recent studies provided new insights into the cellular source, regulation and function of IL-17F. IL-17F/IL-17A expression ratio is higher in psoriatic skin compared to SpA synovitis. IL-17F-expressing T cells produce different proinflammatory mediators than IL-17A-expressing cells, and IL-17F and IL-17A signal through different receptor complex. Dual IL-17A and IL-17F neutralization resulted in greater suppression of downstream inflammatory and tissue remodelling responses. Furthermore, there is additional evidence of IL-23-independent IL-17 production. In contrast to IL-17A, IL-17F and IL-17C, which play proinflammatory roles in skin and joint inflammation, an anti-inflammatory function is proposed for IL-17D. An increase in IL-17E is associated with subclinical gut microbiome alterations after anti-IL-17A therapy in SpA patients.

Summary: IL-17 family cytokines may act as agonists or antagonists to IL-17A contributing in concert to local inflammatory responses. Understanding their function and identifying their cellular sources, and molecular mechanisms driving their expression will be the key to designing rational therapies in SpA.

Trial registration: ClinicalTrials.gov NCT03536884.

Conflict of interest statement

There are no conflicts of interest.

Copyright © 2021 The Author(s). Published by Wolters Kluwer Health, Inc.

Figures

Box 1
Box 1
no caption available
FIGURE 1
FIGURE 1
Recent ex vivo, in vitro and in vivo evidence supporting the role of IL-17 family cytokines in the pathogenesis of SpA. Created with BioRender.com. SpA, spondyloarthritis.

References

    1. Baeten D, Sieper J, Braun J, et al. . Secukinumab, an interleukin-17A inhibitor, in ankylosing spondylitis. N Engl J Med 2015; 373:2534–2548.
    1. McInnes IB, Mease PJ, Kirkham B, et al. . Secukinumab, a human antiinterleukin-17A monoclonal antibody, in patients with psoriatic arthritis (FUTURE 2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2015; 386:1137–1146.
    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.
    1. Mease PJ, Genovese MC, Greenwald MW, et al. . Brodalumab, an anti-IL17RA monoclonal antibody, in psoriatic arthritis. N Engl J Med 2014; 370:2295–2306.
    1. Mease PJ, McInnes IB, Kirkham B, et al. . Secukinumab inhibition of interleukin-17A in Patients with psoriatic arthritis. N Engl J Med 2015; 373:1329–1339.
    1. Mease P, van der Heijde D, Landewe R, et al. . Secukinumab improves active psoriatic arthritis symptoms and inhibits radiographic progression: primary results from the randomised, double-blind, phase III FUTURE 5 study. Ann Rheum Dis 2018; 77:890–897.
    1. Sieper J, Deodhar A, Marzo-Ortega H, et al. . Secukinumab efficacy in anti-TNF-naive and anti-TNF-experienced subjects with active ankylosing spondylitis: results from the MEASURE 2 Study. Ann Rheum Dis 2017; 76:571–592.
    1. Deodhar A, Poddubnyy D, Pacheco-Tena C, et al. . Efficacy and safety of ixekizumab in the treatment of radiographic axial spondyloarthritis: sixteen-week results from a Phase III randomized, double-blind, placebo-controlled trial in patients with prior inadequate response to or intolerance of tumor necrosis factor inhibitors. Arthritis Rheumatol 2019; 71:599–611.
    1. Deodhar A, van der Heijde D, Gensler LS, et al. . Ixekizumab for patients with nonradiographic axial spondyloarthritis (COAST-X): a randomised, placebo-controlled trial. Lancet 2020; 395:53–64.
    1. Dougados M, Wei JC, Landewe R, et al. . Efficacy and safety of ixekizumab through 52 weeks in two phase 3, randomised, controlled clinical trials in patients with active radiographic axial spondyloarthritis (COAST-V and COAST-W). Ann Rheum Dis 2020; 79:176–185.
    1. van der Heijde D, Mease PJ, Landewe RBM, et al. . Secukinumab provides sustained low rates of radiographic progression in psoriatic arthritis: 52-week results from a phase 3 study, FUTURE 5. Rheumatology 2020; 59:1325–1334.
    1. Braun J, Baraliakos X, Deodhar A, et al. . Secukinumab shows sustained efficacy and low structural progression in ankylosing spondylitis: 4-year results from the MEASURE 1 study. Rheumatology 2019; 58:859–868.
    1. Gladman DD, Orbai AM, Klitz U, et al. . Ixekizumab and complete resolution of enthesitis and dactylitis: integrated analysis of two phase 3 randomized trials in psoriatic arthritis. Arthritis Res Ther 2019; 21:38.
    1. Schett G, Baraliakos X, Van den Bosch F, et al. . Secukinumab efficacy on enthesitis in patients with ankylosing spondylitis: pooled analysis of four pivotal Phase 3 studies. J Rheumatol 2021; Mar 15:jrheum.201111. doi: 10.3899/jrheum.201111. [Epub ahead of print].
    1. Thaci D, Blauvelt A, Reich K, et al. . Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate to severe plaque psoriasis: CLEAR, a randomized controlled trial. J Am Acad Dermatol 2015; 73:400–409.
    1. Gordon KB, Colombel JF, Hardin DS. Phase 3 trials of ixekizumab in moderate-to-severe plaque psoriasis. N Engl J Med 2016; 375.
    1. Hueber W, Sands BE, Lewitzky S, et al. . Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn's disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012; 61:1693–1700.
    1. Dick AD, Tugal-Tutkun I, Foster S, et al. . Secukinumab in the treatment of noninfectious uveitis: results of three randomized, controlled clinical trials. Ophthalmology 2013; 120:777–787.
    1. Baeten D, Ostergaard M, Wei JC, et al. . Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-finding phase 2 study. Ann Rheum Dis 2018; 77:1295–1302.
    1. Deodhar A, Gensler LS, Sieper J, et al. . Three multicenter, randomized, double-blind, placebo-controlled studies evaluating the efficacy and safety of ustekinumab in axial spondyloarthritis. Arthritis Rheumatol 2019; 71:258–270.
    1. Sands BE, Sandborn WJ, Panaccione R, et al. . Ustekinumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2019; 381:1201–1214.
    1. Hanzel J, D’Haens GR. Antiinterleukin-23 agents for the treatment of ulcerative colitis. Expert Opin Biol Ther 2020; 20:399–406.
    1. Li H, Chen J, Huang A, et al. . Cloning and characterization of IL-17B and IL-17C, two new members of the IL-17 cytokine family. Proc Natl Acad Sci USA 2000; 97:773–778.
    1. Lee J, Ho WH, Maruoka M, et al. . IL-17E, a novel proinflammatory ligand for the IL-17 receptor homolog IL-17Rh1. J Biol Chem 2001; 276:1660–1664.
    1. Starnes T, Robertson MJ, Sledge G, et al. . Cutting edge: IL-17F, a novel cytokine selectively expressed in activated T cells and monocytes, regulates angiogenesis and endothelial cell cytokine production. J Immunol 2001; 167:4137–4140.
    1. Dubin PJ, Kolls JK. Interleukin-17A and interleukin-17F: a tale of two cytokines. Immunity 2009; 30:9–11.
    1. Zhou L, Littman DR. Transcriptional regulatory networks in Th17 cell differentiation. Curr Opin Immunol 2009; 21:146–152.
    1. Onishi RM, Gaffen SL. Interleukin-17 and its target genes: mechanisms of interleukin-17 function in disease. Immunology 2010; 129:311–321.
    1. Cole S, Murray J, Simpson C, et al. . Interleukin (IL)-12 and IL-18 synergize to promote MAIT cell IL-17A and IL-17F production independently of IL-23 signaling. Front Immunol 2020; 11:585134.
    2. This work demonstrated that innate lymphocytes, including MAIT cells, gd T cells and ILC3s produce predominantly IL-17F cytokine and identified IL-17F-producing cells in psoriatic lesional skin. The authors extended a previous finding that dual blockade of IL-17A and IL-17F is required for optimal inhibition of downstream inflammatory responses. Furthermore, this study provides further evidence towards the contribution of IL-17A and IL-17F to tissue inflammation independently of IL-23, advancing our understanding of divergent results of therapeutic targeting IL-17A and IL-23 in AS.

    1. Kjer-Nielsen L, Patel O, Corbett AJ, et al. . MR1 presents microbial vitamin B metabolites to MAIT cells. Nature 2012; 491:717–723.
    1. Treiner E, Duban L, Bahram S, et al. . Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature 2003; 422:164–169.
    1. Teunissen MBM, Yeremenko NG, Baeten DLP, et al. . The IL-17A-producing CD8+ T-cell population in psoriatic lesional skin comprises mucosa-associated invariant T cells and conventional T cells. J Investig Dermatol 2014; 134:2898–2907.
    1. Menon B, Gullick NJ, Walter GJ, et al. . Interleukin-17+CD8+ T cells are enriched in the joints of patients with psoriatic arthritis and correlate with disease activity and joint damage progression. Arthritis Rheumatol 2014; 66:1272–1281.
    1. Gracey E, Qaiyum Z, Almaghlouth I, et al. . IL-7 primes IL-17 in mucosal-associated invariant T (MAIT) cells, which contribute to the Th17-axis in ankylosing spondylitis. Ann Rheum Dis 2016; 75:2124–2132.
    1. Raychaudhuri SK, Abria C, Mitra A, Raychaudhuri SP. Functional significance of MAIT cells in psoriatic arthritis. Cytokine 2020; 125:154855.
    1. Hayashi E, Chiba A, Tada K, et al. . Involvement of mucosal-associated invariant T cells in ankylosing spondylitis. J Rheumatol 2016; 43:1695–1703.
    1. Toussirot E, Saas P. MAIT cells: potent major cellular players in the IL-17 pathway of spondyloarthritis? RMD Open 2018; 4:e000821.
    1. Cuthbert RJ, Watad A, Fragkakis EM, et al. . Evidence that tissue resident human enthesis gammadeltaT-cells can produce IL-17A independently of IL-23R transcript expression. Ann Rheum Dis 2019; 78:1559–1565.
    1. Webster KE, Kim HO, Kyparissoudis K, et al. . IL-17-producing NKT cells depend exclusively on IL-7 for homeostasis and survival. Mucosal Immunol 2014; 7:1058–1067.
    1. Hassane M, Jouan Y, Creusat F, et al. . Interleukin-7 protects against bacterial respiratory infection by promoting IL-17A-producing innate T-cell response. Mucosal Immunol 2020; 13:128–139.
    1. Yoshiga Y, Goto D, Segawa S, et al. . Invariant NKT cells produce IL-17 through IL-23-dependent and -independent pathways with potential modulation of Th17 response in collagen-induced arthritis. Int J Mol Med 2008; 22:369–374.
    1. Hasegawa E, Sonoda KH, Shichita T, et al. . IL-23-independent induction of IL-17 from gammadeltaT cells and innate lymphoid cells promotes experimental intraocular neovascularization. J Immunol 2013; 190:1778–1787.
    1. Lee JS, Tato CM, Joyce-Shaikh B, et al. . Interleukin-23-independent IL-17 production regulates intestinal epithelial permeability. Immunity 2015; 43:727–738.
    1. Fiechter RH, de Jong HM, van Mens LJJ, et al. . IL-12p40/IL-23p40 blockade with ustekinumab decreases the synovial inflammatory infiltrate through modulation of multiple signaling pathways including MAPK-ERK and Wnt. Front Immunol 2021; 12:611656.
    1. Letarouilly JG, Flachaire B, Labadie C, et al. . Secukinumab and ustekinumab treatment in psoriatic arthritis: results of a direct comparison. Rheumatology (Oxford) 2020; Nov 24:keaa710. doi: 10.1093/rheumatology/keaa710. [Epub ahead of print].
    1. Melton AC, Melrose J, Alajoki L, et al. . Regulation of IL-17A production is distinct from IL-17F in a primary human cell co-culture model of T cell-mediated B cell activation. PLoS One 2013; 8:e58966.
    1. Gomez-Rodriguez J, Sahu N, Handon R, et al. . Differential expression of interleukin-17A and -17F is coupled to T cell receptor signaling via inducible T cell kinase. Immunity 2009; 31:587–597.
    1. Burns LA, Maroof A, Marshall D, et al. . Presence, function, and regulation of IL-17F-expressing human CD4(+) T cells. Eur J Immunol 2020; 50:568–580.
    2. This study provides evidence that IL-17A and IL-17F are differentially regulated upon T cell stimulation and that IL-17F-producing and IL-17A-producing CD4+ T cells display different cytokine profiles. Findings demonstrate that IL-17F contributes to inflammation since dual IL-17A and IL-17F blockade is more effective at reducing inflammatory cytokine production than blockade of IL-17A alone.

    1. Aschenbrenner D, Foglierini M, Jarrossay D, et al. . An immunoregulatory and tissue-residency program modulated by c-MAF in human TH17 cells. Nat Immunol 2018; 19:1126–1136.
    1. Yang XP, Ghoreschi K, Steward-Tharp SM, et al. . Opposing regulation of the locus encoding IL-17 through direct, reciprocal actions of STAT3 and STAT5. Nat Immunol 2011; 12:247–254.
    1. Miller PG, Bonn MB, McKarns SC. Transmembrane TNF-TNFR2 impairs Th17 differentiation by promoting Il2 expression. J Immunol 2015; 195:2633–2647.
    1. Kaaij MH, van Tok MN, Blijdorp IC, et al. . Transmembrane TNF drives osteoproliferative joint inflammation reminiscent of human spondyloarthritis. J Exp Med 2020; 217:e20200288.
    1. Kolbinger F, Loesche C, Valentin MA, et al. . beta-Defensin 2 is a responsive biomarker of IL-17A-driven skin pathology in patients with psoriasis. J Allergy Clin Immunol 2017; 139:923–932. e8.
    1. Glatt S, Baeten D, Baker T, et al. . 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. Ann Rheum Dis 2018; 77:523–532.
    1. Chen S, Blijdorp IC, van Mens LJJ, et al. . Interleukin 17A and IL-17F expression and functional responses in rheumatoid arthritis and peripheral spondyloarthritis. J Rheumatol 2020; 47:1606–1613.
    2. This study demonstrates that IL-17A and IL-17F expression are higher in psoriatic skin compared to the inflamed SpA joint, with a striking inverse IL-17A/IL-17F expression ratio in the joint compared to the skin compartment in SpA. These data suggest that the relative contribution of IL-17F in chronic tissue inflammation may be more prominent in the skin than in joints.

    1. Gordon KB, Foley P, Krueger JG, et al. . Bimekizumab efficacy and safety in moderate to severe plaque psoriasis (BE READY): a multicentre, double-blind, placebo-controlled, randomised withdrawal phase 3 trial. Lancet 2021; 397:475–486.
    2. This phase 3 randomised controlled trial study showed a rapid and substantial response in patients with moderate to severe plaque psoriasis, which was durable over 56 weeks. These data further support the therapeutic value of dual inhibition of IL-17A and IL-17F in PsO.

    1. Reich K, Papp KA, Blauvelt A, et al. . Bimekizumab versus ustekinumab for the treatment of moderate to severe plaque psoriasis (BE VIVID): efficacy and safety from a 52-week, multicentre, double-blind, active comparator and placebo controlled phase 3 trial. Lancet 2021; 397:487–498.
    2. The head-to-head trial evaluated dual IL-17A and IL-17F versus IL-12/IL-23 inhibition in psoriasis over 52 weeks, with results suggesting that bimekizumab is superior to ustekinumab and placebo as shown by significantly higher response rates for all primary and secondary endpoints.

    1. Blauvelt A, Papp KA, Merola JF, et al. . Bimekizumab for patients with moderate to severe plaque psoriasis: 60-week results from BE ABLE 2, a randomized, double-blinded, placebo-controlled, phase 2b extension study. J Am Acad Dermatol 2020; 83:1367–1374.
    1. Papp KA, Merola JF, Gottlieb AB, et al. . Dual neutralization of both interleukin 17A and interleukin 17F with bimekizumab in patients with psoriasis: Results from BE ABLE 1, a 12-week randomized, double-blinded, placebo-controlled phase 2b trial. J Am Acad Dermatol 2018; 79:277–286. e10.
    1. Warren RBA, Bagel J, Papp KA, et al. . Bimekizumab efficacy and safety versus adalimumab in patients with moderate to severe plaque psoriasis: results from a multicenter, randomized, double-blinded active comparator-controlled phase 3 trial (Be Sure). SKIN J Cutan Med 2021; 5:s15.
    2. The head-to-head trial in patients with moderate to severe plaque psoriasis that demonstrated uperiority of bimekizumab to adalimumab over 16 weeks of treatment in terms of the speed, depth and durability of skin clearance.

    1. Blauvelt A, Reich K, Tsai TF, et al. . Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-to-severe plaque psoriasis up to 1 year: Results from the CLEAR study. J Am Acad Dermatol 2017; 76:60–69. e9.
    1. Griffiths CE, Reich K, Lebwohl M, et al. . Comparison of ixekizumab with etanercept or placebo in moderate-to-severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet 2015; 386:541–551.
    1. Ritchlin CT, Kavanaugh A, Merola JF, et al. . Bimekizumab in patients with active psoriatic arthritis: results from a 48-week, randomised, double-blind, placebo-controlled, dose-ranging phase 2b trial. Lancet 2020; 395:427–440.
    1. van der Heijde D, Gensler LS, Deodhar A, et al. . Dual neutralisation of interleukin-17A and interleukin-17F with bimekizumab in patients with active ankylosing spondylitis: results from a 48-week phase IIb, randomised, double-blind, placebo-controlled, dose-ranging study. Ann Rheum Dis 2020; 79:595–604.
    2. This is the first study of bimekizumab in patients with active AS, demonstrating that bimekizumab-treated patients had rapid and significant ASAS40 responses versus placebo at week 12, supported by all secondary efficacy endpoints. These results validate a dual neutralisation of IL-17A and IL-17F with bimekizumab as a novel therapeutic option for the treatment of AS.

    1. Shah M, Maroof A, Gikas P, et al. . Dual neutralisation of IL-17F and IL-17A with bimekizumab blocks inflammation-driven osteogenic differentiation of human periosteal cells. RMD Open 2020; 6:e001306.
    2. This study shows a direct equal effect of IL-17A and IL-17F on progenitor cells from the periosteum. T helper 17 or γδ-T cell supernatants or serum obtained from patients with AS potently stimulated in vitro bone formation, which was blocked deeper by dual inhibition of IL-17A and IL-17F than by neutralisation of IL-17A or IL-17F alone.

    1. Lauffer F, Jargosch M, Baghin V, et al. . IL-17C amplifies epithelial inflammation in human psoriasis and atopic eczema. J Eur Acad Dermatol Venereol 2020; 34:800–809.
    2. This study reported elevated IL-17C in various inflammatory skin diseases and demonstrated that IL-17C amplifies epithelial inflammation and immune cell influx to the skin. IL-17C depletion significantly reduced inflammatory mediators in human skin biopsies of psoriasis ex vivo.

    1. Chiricozzi A, Guttman-Yassky E, Suarez-Farinas M, et al. . Integrative responses to IL-17 and TNF-alpha in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J Investig Dermatol 2011; 131:677–687.
    1. Krueger JG, Wharton KA, Jr, Schlitt T, et al. . IL-17A inhibition by secukinumab induces early clinical, histopathologic, and molecular resolution of psoriasis. J Allergy Clin Immunol 2019; 144:750–763.
    1. Manasson J, Wallach DS, Guggino G, et al. . Interleukin-17 inhibition in spondyloarthritis is associated with subclinical gut microbiome perturbations and a distinctive interleukin-25-driven intestinal inflammation. Arthritis Rheumatol 2020; 72:645–657.
    2. This study reported that IL-17 inhibition-induced subclinical gut inflammation and intestinal dysbiosis in SpA patients is associated with overexpression of IL-25/IL-17E–producing cells, suggesting that IL-17i-induced Crohn's disease might be driven by a specific IL-17 pathway.

    1. Senra L, Stalder R, Alvarez Martinez D, et al. . Keratinocyte-derived IL-17E contributes to inflammation in psoriasis. J Investig Dermatol 2016; 136:1970–1980.
    1. Reynolds JM, Lee YH, Shi Y, et al. . Interleukin-17B antagonizes interleukin-25-mediated mucosal inflammation. Immunity 2015; 42:692–703.
    1. Camelo A, Barlow JL, Drynan LF, et al. . Blocking IL-25 signalling protects against gut inflammation in a type-2 model of colitis by suppressing nuocyte and NKT derived IL-13. J Gastroenterol 2012; 47:1198–1211.
    1. McHenga SS, Wang D, Janneh FM, et al. . Differential dose effects of recombinant IL-25 on the development of dextran sulfate sodium-induced colitis. Inflamm Res 2010; 59:879–887.
    1. Chen SMC, van Tok, Melissa, et al. . Interleukin-17D, a cytokine derived from stromal cells, attenuates joint inflammation. Arthritis Rheumatol 2020; 72: suppl 10: . [Accessed 26 April 2021].
    2. This study demonstrates that expression of IL-17D is localized to synovial stromal cells and inversely correlates with synovial inflammation in SpA. Mice lacking IL-17D. This suggests that IL-17D may exert an anti-inflammatory effect on joint inflammation in SpA and requires further investigation.

    1. Chen S, Paveley R, Kraal L, et al. . Selective targeting of PI3Kdelta suppresses human IL-17-producing T cells and innate-like lymphocytes and may be therapeutic for IL-17-mediated diseases. J Autoimmun 2020; 111:102435.
    2. This publication reports that selective inhibition of PI3Kδ suppresses IL-23-dependent and IL-23-independent production IL-17A and IL-17F by Th17-like cells and innate T cells from a healthy donor and SpA primary cells. This inhibition has a functional impact on the inflammatory and tissue remodelling responses in target cells in psoriasis and SpA. This supports a rationale to target PI3Kδ in IL-17-driven diseases.

    1. O’Connor W, Jr, Kamanaka M, Booth CJ, et al. . A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol 2009; 10:603–609.
    1. Tang C, Kakuta S, Shimizu K, et al. . Suppression of IL-17F, but not of IL-17A, provides protection against colitis by inducing Treg cells through modification of the intestinal microbiota. Nat Immunol 2018; 19:755–765.
    1. Chong WP, Mattapallil MJ, Raychaudhuri K, et al. . The cytokine IL-17A limits Th17 pathogenicity via a negative feedback loop driven by autocrine induction of IL-24. Immunity 2020; 53:384–397. e5.
    1. Goepfert A, Lehmann S, Blank J, et al. . Structural analysis reveals that the cytokine IL-17F forms a homodimeric complex with receptor IL-17RC to drive IL-17RA-independent signaling. Immunity 2020; 52:499–512. e5.
    2. This publication reports the crystal structure of the extracellular domain of human IL-17RC in complex with IL-17F, providing a structural basis for IL-17F signalling through IL-17RC and suggesting the possibility of IL-17RA-independent IL-17 signalling pathways.

    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; 111:1599–1607.

Source: PubMed

3
Suscribir