IL-23 blockade with guselkumab potentially modifies psoriasis pathogenesis: rationale and study protocol of a phase 3b, randomised, double-blind, multicentre study in participants with moderate-to-severe plaque-type psoriasis (GUIDE)

Kilian Eyerich, Peter Weisenseel, Andreas Pinter, Knut Schäkel, Khusru Asadullah, Sven Wegner, Ernesto J Muñoz-Elias, Holger Bartz, Friedmann J H Taut, Kristian Reich, Kilian Eyerich, Peter Weisenseel, Andreas Pinter, Knut Schäkel, Khusru Asadullah, Sven Wegner, Ernesto J Muñoz-Elias, Holger Bartz, Friedmann J H Taut, Kristian Reich

Abstract

Background: Guselkumab is an interleukin (IL)-23 pathway blocker with proven efficacy in patients with moderate-to-severe plaque psoriasis. Early intervention with guselkumab may result in changes to the clinical disease course versus later intervention.

Methods and analysis: Here we present the rationale and design of a phase 3b, randomised, double-blind, multicentre study (GUIDE), comparing treatment effects of guselkumab in patients with short (≤2 years) or longer (>2 years) duration of plaque-type psoriasis, measured from first appearance of psoriatic plaques. Participants achieving skin clearance (Psoriasis Area and Severity Index (PASI)=0) by week 20 and maintaining complete clearance at week 28 visit ('super-responders' (SRe)) will be randomised to continue approved maintenance dosing every 8 weeks (q8w) versus an investigational maintenance dosing interval of 16 weeks (q16w) until week 68. Primary endpoint: proportion of participants in the q8w vs q16w arms with absolute PASI <3 at week 68. Participants with PASI <3 at week 68 will be withdrawn from guselkumab treatment for up to 48 weeks. Participants not achieving SRe criteria (non-SRe) will remain in the study with q8w guselkumab dosing through week 68. Additional to serum samples obtained from all patients, skin biopsies and whole-blood samples will be taken from SRe and non-SRe participants at various time points in optional substudies. Analyses include: genetics; immunophenotyping (fluorescence-activated cell sorting); gene and protein expression profiling; immunohistology. By merging clinical endpoints with mechanistic findings, this study aims to elucidate how IL-23 blockade with guselkumab can modify the disease course by altering molecular and cellular drivers that cause relapse after treatment withdrawal, particularly among SRe.

Ethics and dissemination: Approval obtained from ethics committee Medical Council Hamburg, Germany (PVN5925). GUIDE is compliant with the Declaration of Helsinki.

Trial registration number: Registered at ClinicalTrials.gov (NCT03818035). All primary endpoint results (prespecified analyses) will be submitted to peer-reviewed, international journals within 18 months after primary completion date.

Keywords: adult dermatology; protocols & guidelines; psoriasis.

Conflict of interest statement

Competing interests: KE reports grants and personal fees from Janssen during the conduct of the study, grants from AbbVie, LEO Pharma, UCB and Novartis, and personal fees from AbbVie, Almirall, BMS, LEO Pharma, Lilly, Sanofi, UCB, Galderma and Novartis outside the submitted work; PW reports receiving honoraria as consultant or speaker from the following companies involved in the development or distribution of drugs for psoriasis: AbbVie, Almirall, Biogen, Celgene, Eli Lilly, Janssen, LEO Pharma, Medac and Novartis, and honoraria received by his institution for active participation in clinical studies sponsored by Janssen, AbbVie and Eli Lilly; AP has no conflicts of interest to report; KS reports conducting clinical studies during the past 36 months with the following companies: AbbVie, Almirall, Boehringer, Celgene, Chugai, Galapagos, Galderma, Janssen-Cilag, LEO Pharma, Lilly, Merck Sharp & Dohme Corp., Novartis Regeneron and UCB Pharma; KA reports honoraria for participation in advisory boards, consultation, clinical trials or as speaker from AbbVie, Almirall, Antabio, Bayer, BMS, Euroimmune, Emphasis, Emeritipharma, Galderma, Janssen, La Roche-Posay, LEO Pharma, L’Oréal, Novartis, Parexel International, Pierre Fabre, Roxall, RG, Sanofi Genzyme, TFS Trial Form Support and UCB; SW is a full-time employee of Janssen-Cilag Germany; EJM-E is a full-time employee of Johnson & Johnson, and is listed as an inventor on a patent application related to uses of guselkumab to treat psoriasis, pending; HB reports personal fees from Janssen-Cilag Germany during the conduct of the study, and personal fees from Janssen-Cilag Germany outside the submitted work; FJHT reports personal fees from Janssen-Cilag, Germany during the conduct of the study and personal fees from Janssen-Cilag, Germany outside the submitted work; KR reports grants and personal fees from Janssen during the conduct of the study, grants from AbbVie, Lilly, LEO Pharma, UCB, Pfizer, Affibody, Biogen-Idec, Boehringer Ingelheim Pharma, BMS, Celgene, Covagen, Forward Pharma, Fresenius Medical Care, Galapagos, Kyowa Kirin, Medac, Merck Sharp & Dohme, Milteny, Novartis, Ocean Pharma, Sandoz, Sanofi, Sun Pharma, Takeda and XBiotech; personal fees from AbbVie, Lilly, LEO Pharma, UCB, Pfizer, Amgen, Affibody, Biogen-Idec, Boehringer Ingelheim Pharma, BMS, Celgene, Covagen, Forward Pharma, GSK, Kyowa Kirin, Medac, Merck Sharp & Dohme Corp, Novartis, Ocean Pharma, Samsung Bioepis, Sandoz, Sanofi, Takeda, Valeant and Xenoport outside the submitted work.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Current pathogenic model of psoriasis. (A) T cell subsets in psoriasis, their differentiation, function and phenotype. High TGF-β concentration promotes IL-10 and IL-17 producing Treg differentiation, while the presence of IL-6, TGF-β, IL-1-β and IL-21 promote dominant Th17 cell differentiation expressing an IL-17/IFN-γ cytokine signature. (B) In early psoriasis, environmental stimuli in combination with a loss of tolerance activate the innate immune system, leading to the production of IL-23 by dermal DCs and macrophages. IL-23 then drives activation and expansion of T17 cells which subsequently generate a cytokine milieu that promotes a feedforward inflammatory cascade in epidermal keratinocytes, leading to parakeratosis and psoriatic lesions. IL-17 autoamplifies its signal by triggering the production of chemokines by activated keratinocytes, which subsequently recruits more T17 cells and other immune cells (eg, neutrophils, DCs and macrophages). Persistent high levels of IL-23 in psoriatic skin sustain IL-17 production, thus fuelling the self-amplifying inflammatory process. As psoriasis progresses to a more chronic state, sustained high levels of IL-23 in combination with low concentrations of TGF-β promotes IL-23R expression, favouring T17 cell differentiation and supressing Treg differentiation. TRM are a subset of non-circulating memory T cells that persist long-term in peripheral tissues and are characterised by the markers CD69 and CD103. (C) Inhibition of the regulatory cytokine IL-23 is hypothesised to lead to long-lasting therapeutic effects by restoring a ‘physiological’ Treg/TRM balance. This is in contrast to the blockade of an effector cytokine, which leads to reduction of inflammatory cells but has less effect on the relative numbers of proinflammatory and anti-inflammatory T cells. Ahr, aryl hydrocarbon receptor; CCR, chemokine receptor; CD, cluster of differentiation; CXCR, CXC chemokine receptor; DC, dendritic cell; IFN, interferon; IL, interleukin; RORγt, retinoic acid receptor-related orphan receptor transcription factor; T17, IL-17 producing T cells; T-bet, T box protein expressed in T cells; TGF, transforming growth factor; Th17, T helper 17; Treg, regulatory T cell; TRM, tissue-resident memory T cell.
Figure 2
Figure 2
Study setup and design. SRe are defined as participants who receive on-label GUS treatment until W20 and respond with a score of PASI=0 at W20 and W28. *Blinded treatment. Group 1: All participants who are enrolled and scheduled to receive GUS 100 mg at W0, W4, then q8w until W28 (study part 1). Group 2a: SRe (PASI score=0 at W20 and W28) randomised to GUS 100 mg q8w at W28–W60 (study part 2). Group 2b: SRe randomised to GUS 100 mg q16w at W28–W60 (study part 2). Group 2c: non-SRe with a PASI score >0 at W20 and/or W28 who will receive GUS 100 mg q8w at W28–W60 (study part 2). Group 2d: SRe with loss of disease control between W28 and W68, who will enter the retreatment arm and receive GUS 100 mg at R0, R8 and R16 calculated from the date of loss of disease control (study part 2). Group 3a: SRe randomised to GUS 100 mg q8w in study part 2 with withdrawal of GUS at W68 (study part 3). Group 3b: SRe randomised to GUS 100 mg q16w in study part 2 with withdrawal of GUS at W68 (study part 3). Group 3c: SRe with fluctuating disease (PASI score 3–5) at W68 or loss of disease control (PASI score >5) at any other visit after W68, who will enter the retreatment arm and receive GUS 100 mg at R0, R8 and R16 calculated from the date of loss of disease control (study part 3). GUS, guselkumab; PASI, Psoriasis Area and Severity Index score; q8w, every 8 weeks; q16w, every 16 weeks; R, retreatment week; SRe, super-responder; W, week.

References

    1. Takeshita J, Grewal S, Langan SM, et al. . Psoriasis and comorbid diseases: epidemiology. J Am Acad Dermatol 2017;76:377–90. 10.1016/j.jaad.2016.07.064
    1. Hawkes JE, Yan BY, Chan TC, et al. . Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol 2018;201:1605–13. 10.4049/jimmunol.1800013
    1. Kim J, Krueger JG. Highly effective new treatments for psoriasis target the IL-23/type 17 T cell autoimmune axis. Annu Rev Med 2017;68:255–69. 10.1146/annurev-med-042915-103905
    1. World Health Organization . Global report on psoriasis, 2016. Available:
    1. Mrowietz U, de Jong EMGJ, Kragballe K, et al. . A consensus report on appropriate treatment optimization and transitioning in the management of moderate-to-severe plaque psoriasis. J Eur Acad Dermatol Venereol 2014;28:438–53. 10.1111/jdv.12118
    1. Blome C, Gosau R, Radtke MA, et al. . Patient-relevant treatment goals in psoriasis. Arch Dermatol Res 2016;308:69–78. 10.1007/s00403-015-1613-8
    1. Mrowietz U, Kragballe K, Reich K, et al. . Definition of treatment goals for moderate to severe psoriasis: a European consensus. Arch Dermatol Res 2011;303:1–10. 10.1007/s00403-010-1080-1
    1. Chan TC, Hawkes JE, Krueger JG. Interleukin 23 in the skin: role in psoriasis pathogenesis and selective interleukin 23 blockade as treatment. Ther Adv Chronic Dis 2018;9:111–9. 10.1177/2040622318759282
    1. Schäkel K, Schön MP, Ghoreschi K. Pathogenese der Psoriasis vulgaris [Pathogenesis of psoriasis vulgaris]. Der Hautarzt 2016;67:422–31.
    1. Chiricozzi A, Romanelli P, Volpe E, et al. . Scanning the immunopathogenesis of psoriasis. Int J Mol Sci 2018;19:179. 10.3390/ijms19010179
    1. Girolomoni G, Strohal R, Puig L, et al. . The role of IL-23 and the IL-23/TH17 immune axis in the pathogenesis and treatment of psoriasis. J Eur Acad Dermatology Venereol 2017;31:1616–26.
    1. Bovenschen HJ, van de Kerkhof PC, van Erp PE, et al. . Foxp3+ regulatory T cells of psoriasis patients easily differentiate into IL-17A-producing cells and are found in lesional skin. J Invest Dermatol 2011;131:1853–60. 10.1038/jid.2011.139
    1. Kannan AK, Su Z, Gauvin DM, et al. . IL-23 induces regulatory T cell plasticity with implications for inflammatory skin diseases. Sci Rep 2019;9:17675. 10.1038/s41598-019-53240-z
    1. Soler DC, McCormick TS. The dark side of regulatory T cells in psoriasis. J Invest Dermatol 2011;131:1785–6. 10.1038/jid.2011.200
    1. Yang L, Li B, Dang E, et al. . Impaired function of regulatory T cells in patients with psoriasis is mediated by phosphorylation of STAT3. J Dermatol Sci 2016;81:85–92. 10.1016/j.jdermsci.2015.11.007
    1. Cheuk S, Wikén M, Blomqvist L, et al. . Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. J Immunol 2014;192:3111–20. 10.4049/jimmunol.1302313
    1. Clark RA. Resident memory T cells in human health and disease. Sci Transl Med 2015;7:269rv1. 10.1126/scitranslmed.3010641
    1. Reich K, Papp KA, Armstrong AW, et al. . Safety of guselkumab in patients with moderate-to-severe psoriasis treated through 100 weeks: a pooled analysis from the randomized VOYAGE 1 and VOYAGE 2 studies. Br J Dermatol 2019;180:1039–49. 10.1111/bjd.17454
    1. Reich K, Griffiths CEM, Gordon KB, et al. . Maintenance of clinical response and consistent safety profile with up to 3 years of continuous treatment with guselkumab: Results from the VOYAGE 1 and VOYAGE 2 trials. J Am Acad Dermatol 2020;82:936–45. 10.1016/j.jaad.2019.11.040
    1. Ghilardi N, Kljavin N, Chen Q, et al. . Compromised humoral and delayed-type hypersensitivity responses in IL-23-deficient mice. J Immunol 2004;172:2827–33. 10.4049/jimmunol.172.5.2827
    1. Gooderham MJ, Papp KA, Lynde CW. Shifting the focus – the primary role of IL-23 in psoriasis and other inflammatory disorders. J Eur Acad Dermatology Venereol 2018;32:1111–9.
    1. Di Meglio P, Nestle FO. The role of IL-23 in the immunopathogenesis of psoriasis. F1000 Biol Rep 2010;2:40. 10.3410/B2-40
    1. Duvallet E, Semerano L, Assier E, et al. . Interleukin-23: a key cytokine in inflammatory diseases. Ann Med 2011;43:503–11. 10.3109/07853890.2011.577093
    1. Puig L. The role of IL 23 in the treatment of psoriasis. Expert Rev Clin Immunol 2017;13:525–34. 10.1080/1744666X.2017.1292137
    1. Sun R, Hedl M, Abraham C. IL23 induces IL23R recycling and amplifies innate receptor-induced signalling and cytokines in human macrophages, and the IBD-protective IL23R R381Q variant modulates these outcomes. Gut 2020;69:264–73. 10.1136/gutjnl-2018-316830
    1. Longman RS, Diehl GE, Victorio DA, et al. . CX₃CR1⁺ mononuclear phagocytes support colitis-associated innate lymphoid cell production of IL-22. J Exp Med 2014;211:1571–83. 10.1084/jem.20140678
    1. Gaffen SL, Jain R, Garg AV, et al. . The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014;14:585–600. 10.1038/nri3707
    1. Durant L, Watford WT, Ramos HL, et al. . Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity 2010;32:605–15. 10.1016/j.immuni.2010.05.003
    1. Ivanov II, McKenzie BS, Zhou L, et al. . The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006;126:1121–33. 10.1016/j.cell.2006.07.035
    1. Cheuk S, Schlums H, Gallais Sérézal I, et al. . CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 2017;46:287–300. 10.1016/j.immuni.2017.01.009
    1. Park CO, Kupper TS. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat Med 2015;21:688–97. 10.1038/nm.3883
    1. Clark RA. Gone but not forgotten: lesional memory in psoriatic skin. J Invest Dermatol 2011;131:283–5. 10.1038/jid.2010.374
    1. Clark RA. Skin-resident T cells: the ups and downs of on site immunity. J Invest Dermatol 2010;130:362–70. 10.1038/jid.2009.247
    1. Wu H, Liao W, Li Q, et al. . Pathogenic role of tissue-resident memory T cells in autoimmune diseases. Autoimmun Rev 2018;17:906–11. 10.1016/j.autrev.2018.03.014
    1. Bhushan M, Bleiker TO, Ballsdon AE, et al. . Anti-E-selectin is ineffective in the treatment of psoriasis: a randomized trial. Br J Dermatol 2002;146:824–31. 10.1046/j.1365-2133.2002.04743.x
    1. Boyman O, Conrad C, Tonel G, et al. . The pathogenic role of tissue-resident immune cells in psoriasis. Trends Immunol 2007;28:51–7. 10.1016/j.it.2006.12.005
    1. Boyman O, Hefti HP, Conrad C, et al. . Spontaneous development of psoriasis in a new animal model shows an essential role for resident T cells and tumor necrosis factor-alpha. J Exp Med 2004;199:731–6. 10.1084/jem.20031482
    1. Suárez-Fariñas M, Fuentes-Duculan J, Lowes MA, et al. . Resolved psoriasis lesions retain expression of a subset of disease-related genes. J Invest Dermatol 2011;131:391–400. 10.1038/jid.2010.280
    1. Gallais Sérézal I, Hoffer E, Ignatov B, et al. . A skewed pool of resident T cells triggers psoriasis-associated tissue responses in never-lesional skin from patients with psoriasis. J Allergy Clin Immunol 2019;143:1444–54. 10.1016/j.jaci.2018.08.048
    1. Kurihara K, Fujiyama T, Phadungsaksawasdi P, et al. . Significance of IL-17A-producing CD8+CD103+ skin resident memory T cells in psoriasis lesion and their possible relationship to clinical course. J Dermatol Sci 2019;95:21–7. 10.1016/j.jdermsci.2019.06.002
    1. Matos TR, O’Malley JT, Lowry EL, et al. . Clinically resolved psoriatic lesions contain psoriasis-specific IL-17-producing αβ T cell clones. J Clin Invest 2017;127:4031–41. 10.1172/JCI93396
    1. Casciano F, Pigatto PD, Secchiero P, et al. . T cell hierarchy in the pathogenesis of psoriasis and associated cardiovascular comorbidities. Front Immunol 2018;9:1390. 10.3389/fimmu.2018.01390
    1. Mehta H, Mashiko S, Angsana J. Differential changes in inflammatory mononuclear phagocyte and T cell profiles within psoriatic skin during treatment with Guselkumab versus Secukinumab. J Invest Dermatol 2021;141:1707–18.
    1. Ali N, Rosenblum MD. Regulatory T cells in skin. Immunology 2017;152:372–81. 10.1111/imm.12791
    1. Lee GR. The balance of Th17 versus Treg cells in autoimmunity. Int J Mol Sci 2018;19:730. 10.3390/ijms19030730
    1. Zhou X, Bailey-Bucktrout SL, Jeker LT, et al. . Instability of the transcription factor FOXP3 leads to the generation of pathogenic memory T cells in vivo. Nat Immunol 2009;10:1000–7. 10.1038/ni.1774
    1. Chen X, Oppenheim JJ. Th17 cells and Tregs: unlikely allies. J Leukoc Biol 2014;95:723–31. 10.1189/jlb.1213633
    1. Van Herck MA, Weyler J, Kwanten WJ, et al. . The differential roles of T cells in non-alcoholic fatty liver disease and obesity. Front Immunol 2019;10:82. 10.3389/fimmu.2019.00082
    1. Sanchez Rodriguez R, Pauli ML, Neuhaus IM, et al. . Memory regulatory T cells reside in human skin. J Clin Invest 2014;124:1027–36. 10.1172/JCI72932
    1. Sugiyama H, Gyulai R, Toichi E, et al. . Dysfunctional blood and target tissue CD4+CD25high regulatory T cells in psoriasis: mechanism underlying unrestrained pathogenic effector T cell proliferation. J Immunol 2005;174:164–73. 10.4049/jimmunol.174.1.164
    1. Aletaha D, Smolen JS. The definition and measurement of disease modification in inflammatory rheumatic diseases. Rheum Dis Clin North Am 2006;32:9–44. 10.1016/j.rdc.2005.09.005
    1. Danese S, Fiorino G, Peyrin-Biroulet L. Early intervention in Crohn’s disease: towards disease modification trials. Gut 2017;66:2179–87. 10.1136/gutjnl-2017-314519
    1. Girolomoni G, Griffiths CEM, Krueger J, et al. . Early intervention in psoriasis and immune-mediated inflammatory diseases: a hypothesis paper. J Dermatolog Treat 2015;26:103–12. 10.3109/09546634.2014.880396
    1. Nast A, Boehncke W-H, Mrowietz U. S3-Leitlinie zur Therapie der Psoriasis vulgaris Update 2011 [S3-guidelines for the treatment of psoriasis vulgaris update 2011]. J Dtsch Dermatol Ges 2011;9:S1–104.
    1. van den Reek JMPA, Seyger MMB, van Lümig PPM, et al. . The journey of adult psoriasis patients towards biologics: past and present - Results from the BioCAPTURE registry. J Eur Acad Dermatol Venereol 2018;32:615–23. 10.1111/jdv.14684
    1. Blauvelt A, Papp KA, Griffiths CEM, et al. . Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: results from the phase III, double-blinded, placebo- and active comparator-controlled voyage 1 trial. J Am Acad Dermatol 2017;76:405–17. 10.1016/j.jaad.2016.11.041
    1. Papp KA, Blauvelt A, Kimball AB, et al. . Patient-reported symptoms and signs of moderate-to-severe psoriasis treated with guselkumab or adalimumab: results from the randomized voyage 1 trial. J Eur Acad Dermatol Venereol 2018;32:1515–22. 10.1111/jdv.14910
    1. Reich K, Armstrong AW, Foley P, et al. . Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: results from the phase III, double-blind, placebo- and active comparator-controlled voyage 2 trial. J Am Acad Dermatol 2017;76:418–31. 10.1016/j.jaad.2016.11.042
    1. Griffiths CEM, Papp KA, Song M, et al. . Maintenance of response through 5 years of continuous guselkumab treatment: results from the phase 3 VOYAGE 1 trial. Poster presented at the 16th annual coastal dermatology symposium, October 15–16 2020.
    1. Gordon K, Armstrong A, Foley Y. Long-term efficacy of guselkumab treatment after drug withdrawal and retreatment in patients with moderate to severe plaque psoriasis: results from VOYAGE 2. presented at AAD annual meeting, 16–18 February Abstract 6748 2018.
    1. Griffiths CE, Papp K, Kimball AB. Two-year efficacy and safety of guselkumab for treatment of moderate-to-severe psoriasis: phase 3 VOYAGE 1 trial (Abstract AB0912). Ann Rheum Dis 2018;77:1580.
    1. Gordon KB, Armstrong AW, Foley P, et al. . Guselkumab efficacy after withdrawal is associated with suppression of serum IL-23-regulated IL-17 and IL-22 in psoriasis: VOYAGE 2 study. J Invest Dermatol 2019;139:2437–46. 10.1016/j.jid.2019.05.016
    1. Liu X, Branigan P, Chen Y, et al. . Identification of clinical and biomarker parameters associated with long-term maintenance of PASI 90 response following guselkumab treatment-withdrawal in psoriasis. Poster presented at the 27th EADV Congress, 12–16 September 2018, Paris, France Abstract:P1894 2018.
    1. Reich K, Armstrong AW, Langley RG, et al. . Guselkumab versus secukinumab for the treatment of moderate-to-severe psoriasis (eclipse): results from a phase 3, randomised controlled trial. Lancet 2019;394:831–9. 10.1016/S0140-6736(19)31773-8
    1. Ghoreschi K, Laurence A, Yang XP, et al. . T helper 17 cell heterogeneity and pathogenicity in autoimmune disease. Trends Immunol 2011;32:395–401. 10.1126/scitranslmed.3010302

Source: PubMed

Подписаться