First-In-Human, Phase 1, Randomized, Dose-Escalation Trial with Recombinant Anti-IL-20 Monoclonal Antibody in Patients with Psoriasis

Alice B Gottlieb, James G Krueger, Mia Sandberg Lundblad, Marie Göthberg, Brett E Skolnick, Alice B Gottlieb, James G Krueger, Mia Sandberg Lundblad, Marie Göthberg, Brett E Skolnick

Abstract

Background: The current trial was a first-in-human clinical trial evaluating the safety, tolerability, pharmacokinetics, pharmacodynamics, and preliminary efficacy of the recombinant monoclonal anti-interleukin-20 (IL-20) antibody, NNC0109-0012, which targets the inflammatory cytokine IL-20.

Methods: In total, 48 patients aged 18 to 75 years with moderate to severe stable chronic plaque psoriasis with affected body surface area ≥15% and physician global assessment score ≥3 were enrolled in this randomized, double-blind, multicenter, placebo-controlled, phase 1 dose-escalation trial. Patients were randomized within each single dose cohort (0.01, 0.05, 0.2, 0.6, 1.5, or 3.0 mg/kg) or multiple dose cohort (0.05, 0.2, 0.5, 1.0, or 2.0 mg/kg; 1 dose every other week for 7 weeks) of NNC0109-0012 or placebo in a 3:1 ratio. In the expansion phase, 7 patients were randomized to weekly doses of 2.0 mg/kg NNC0109-0012 or placebo for 7 weeks. The primary objective, safety and tolerability, was assessed by evaluating adverse events (AEs). Additional endpoints included pharmacokinetics, pharmacodynamics, and clinical response (assessed using the Psoriasis Area and Severity Index [PASI] score).

Results: AEs were reported in 85% of patients (n = 40) in the initial study phases (NNC0109-0012, 83%; placebo, 92%) and in 4 of 7 patients in the multiple-dose expansion phase. One serious AE was reported but was judged not to be causally related to NNC0109-0012. No dose-limiting toxicities were reported. NNC0109-0012 pharmacokinetics was similar to other monoclonal antibodies, with an average half-life of approximately 3 weeks. There was a dose-proportional increase in area under the curve and maximum concentration after single dosing. No substantial changes in pharmacodynamic parameters were observed. The expansion phase was terminated early due to apparent lack of PASI improvement.

Conclusion: Single and multiple doses of NNC0109-0012, ranging from 0.05 to 3.0 mg/kg, were well tolerated in patients with psoriasis and exhibited pharmacokinetics similar to that of other monoclonal antibodies.

Trial registration: ClinicalTrials.gov NCT01261767.

Conflict of interest statement

Competing Interests: The authors have the following interests: The study was funded by Novo Nordisk A/S, Denmark. Writing support was provided by Heather Heerssen, PhD, Jane Phillips, PhD, and Maryann Travaglini, PharmD, of Complete Healthcare Communications, Inc. (Chadds Ford, PA, USA). This support was funded by Novo Nordisk. A Gottlieb: Consulting/advisory board: Amgen Inc., Astellas, Akros, Centocor (Janssen), Inc., Celgene Corp., Bristol Myers Squibb Co., Beiersdorf, Inc., Abbott Labs. (AbbVie), TEVA, Actelion, UCB, Novo Nordisk, Novartis, Dermipsor Ltd., Incyte, Pfizer, Canfite, Lilly, Coronado, Vertex, Karyopharm, CSL Behring Biotherapies for Life, GlaxoSmithKline, Xenoport, Catabasis, Sanofi Aventis; DUSA; Research/educational grants (paid to Tufts Medical Center): Centocor (Janssen), Amgen, Abbott (AbbVie), Novartis, Celgene, Pfizer, Lilly, Coronado, Levia, Merck, Xenoport. J Krueger: Consulting/advisory board: Abbvie, Akros, Amgen, Biogen-Idec, Bristol Meyers Squibb, Boehringer-Ingelheim, Centocor/Janssen, Celgene, Delenex, Dermira, Leo Pharma, Eli Lilly, GlaxoSmithKline, Glenmark, Merck, Novartis, Novo Nordisk, Pfizer, Xenoport. M Sandberg Lundblad: Employee and stockholder of Novo Nordisk. M Gothberg: Employee and stockholder of Novo Nordisk. B Skolnick: At the time of manuscript preparation, Dr Skolnick was an employee and stockholder of Novo Nordisk. He is now employed with Cushing Neuroscience Institute, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY. NNC0109-0012 has been patented; however, there are no products in development or marketed products to declare. This does not alter our adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Fig 1. Consort flow diagram.
Fig 1. Consort flow diagram.
Patient disposition in (A) the single-dose and multiple-dose dose-escalation phases and (B) the multiple-dose expansion phase. Represents the safety analysis set. AE = adverse event.
Fig 2. Study design.
Fig 2. Study design.
The first dose in the multiple-dose dose-escalation phase was administered after the third dose level of the single-dose dose-escalation phase (0.2 mg/kg) was evaluated by a study safety group; thereafter, the single- and multiple-dose dose-escalation phases were performed in parallel.
Fig 3. Mean anti‒IL-20 serum concentration-time profiles.
Fig 3. Mean anti‒IL-20 serum concentration-time profiles.
Mean serum concentration-time profiles following (A) single dosing or (B) multiple dosing in the dose-escalation phase and individual anti‒IL-20 serum concentration-time profiles following multiple dosing in (C) the expansion phase on a semi-logarithmic scale in patients with psoriasis.
Fig 4. Mean PASI total score by…
Fig 4. Mean PASI total score by visit.
Mean PASI total score by visit following (A) single dosing or (B) multiple dosing in the dose-escalation phase and (C) multiple dosing in the expansion phase. PASI = Psoriasis Area and Severity Index.
Fig 5. Histology image from a single…
Fig 5. Histology image from a single patient.
Patient from the expansion phase treated with anti‒IL-20. By week 15, this patient experienced marked improvement in epidermal hyperplasia and acanthosis, a large reduction in keratin 16, associated reductions in proliferating keratinocytes (Ki67), and marked reductions in CD11c+ dendritic cells. CD11c = integrin alpha X chain protein; H&E = hematoxylin and eosin stain; K16 = keratin 16; Ki67 = cellular marker for proliferation; NL = nonlesional (skin); LS = longitudinal section.

References

    1. Lebwohl M. Psoriasis. Lancet. 2003;361(9364):1197–204.
    1. Asadullah K, Volk HD, Sterry W. Novel immunotherapies for psoriasis. Trends Immunol. 2002;23(1):47–53.
    1. Gottlieb AB. Psoriasis: emerging therapeutic strategies. Nat Rev Drug Discov. 2005;4(1):19–34.
    1. Gelfand JM, Stern RS, Nijsten T, Feldman SR, Thomas J, Kist J, et al. The prevalence of psoriasis in African Americans: results from a population-based study. J Am Acad Dermatol. 2005;52(1):23–6.
    1. Robinson D Jr, Hackett M, Wong J, Kimball AB, Cohen R, Bala M, et al. Co-occurrence and comorbidities in patients with immune-mediated inflammatory disorders: an exploration using US healthcare claims data, 2001–2002. Curr Med Res Opin. 2006;22(5):989–1000.
    1. Stern RS, Nijsten T, Feldman SR, Margolis DJ, Rolstad T. Psoriasis is common, carries a substantial burden even when not extensive, and is associated with widespread treatment dissatisfaction. J Investig Dermatol Symp Proc. 2004;9(2):136–9.
    1. Kurd SK, Gelfand JM. The prevalence of previously diagnosed and undiagnosed psoriasis in US adults: results from NHANES 2003–2004. J Am Acad Dermatol. 2009;60(2):218–24. 10.1016/j.jaad.2008.09.022
    1. Icen M, Crowson CS, McEvoy MT, Dann FJ, Gabriel SE, Maradit Kremers H. Trends in incidence of adult-onset psoriasis over three decades: a population-based study. J Am Acad Dermatol. 2009;60(3):394–401. 10.1016/j.jaad.2008.10.062
    1. Setty AR, Curhan G, Choi HK. Obesity, waist circumference, weight change, and the risk of psoriasis in women: Nurses' Health Study II. Arch Intern Med. 2007;167(15):1670–5.
    1. Novartis announces FDA approval for first IL-17A antagonist Cosentyx (secukinumab) for moderate-to-severe plaque psoriasis patients 2015. Available: . Accessed: 12 March 2015.
    1. Leman J, Burden AD. Sequential use of biologics in the treatment of moderate-to-severe plaque psoriasis. Br J Dermatol. 2012;167 suppl 3:12–20. 10.1111/j.1365-2133.2012.11209.x
    1. Hofmann SR, Rosen-Wolff A, Tsokos GC, Hedrich CM. Biological properties and regulation of IL-10 related cytokines and their contribution to autoimmune disease and tissue injury. Clin Immunol. 2012;143(2):116–27. 10.1016/j.clim.2012.02.005
    1. Kunz S, Wolk K, Witte E, Witte K, Doecke WD, Volk HD, et al. Interleukin (IL)-19, IL-20 and IL-24 are produced by and act on keratinocytes and are distinct from classical ILs. Exp Dermatol. 2006;15(12):991–1004.
    1. Romer J, Hasselager E, Norby PL, Steiniche T, Thorn Clausen J, Kragballe K. Epidermal overexpression of interleukin-19 and -20 mRNA in psoriatic skin disappears after short-term treatment with cyclosporine a or calcipotriol. J Invest Dermatol. 2003;121(6):1306–11.
    1. Wei CC, Chen WY, Wang YC, Chen PJ, Lee JY, Wong TW, et al. Detection of IL-20 and its receptors on psoriatic skin. Clin Immunol. 2005;117(1):65–72.
    1. Otkjaer K, Kragballe K, Funding AT, Clausen JT, Noerby PL, Steiniche T, et al. The dynamics of gene expression of interleukin-19 and interleukin-20 and their receptors in psoriasis. Br J Dermatol. 2005;153(5):911–8.
    1. Wolk K, Witte K, Witte E, Proesch S, Schulze-Tanzil G, Nasilowska K, et al. Maturing dendritic cells are an important source of IL-29 and IL-20 that may cooperatively increase the innate immunity of keratinocytes. J Leukoc Biol. 2008;83(5):1181–93. 10.1189/jlb.0807525
    1. Wolk K, Kunz S, Asadullah K, Sabat R. Cutting edge: immune cells as sources and targets of the IL-10 family members? J Immunol. 2002;168(11):5397–402.
    1. Sa SM, Valdez PA, Wu J, Jung K, Zhong F, Hall L, et al. The effects of IL-20 subfamily cytokines on reconstituted human epidermis suggest potential roles in cutaneous innate defense and pathogenic adaptive immunity in psoriasis. J Immunol. 2007;178(4):2229–40.
    1. Dumoutier L, Leemans C, Lejeune D, Kotenko SV, Renauld JC. Cutting edge: STAT activation by IL-19, IL-20 and mda-7 through IL-20 receptor complexes of two types. J Immunol. 2001;167(7):3545–9.
    1. Wolk K, Haugen HS, Xu W, Witte E, Waggie K, Anderson M, et al. IL-22 and IL-20 are key mediators of the epidermal alterations in psoriasis while IL-17 and IFN-gamma are not. J Mol Med (Berl). 2009;87(5):523–36.
    1. Blumberg H, Conklin D, Xu WF, Grossmann A, Brender T, Carollo S, et al. Interleukin 20: discovery, receptor identification, and role in epidermal function. Cell. 2001;104(1):9–19.
    1. Tohyama M, Hanakawa Y, Shirakata Y, Dai X, Yang L, Hirakawa S, et al. IL-17 and IL-22 mediate IL-20 subfamily cytokine production in cultured keratinocytes via increased IL-22 receptor expression. Eur J Immunol. 2009;39(10):2779–88. 10.1002/eji.200939473
    1. Stenderup K, Rosada C, Worsaae A, Dagnaes-Hansen F, Steiniche T, Hasselager E, et al. Interleukin-20 plays a critical role in maintenance and development of psoriasis in the human xenograft transplantation model. Br J Dermatol. 2009;160(2):284–96. 10.1111/j.1365-2133.2008.08890.x
    1. Lundblad MS, Overgaard RV, Gothberg M, Fjording MS, Watson E. Clinical pharmacokinetics of the anti-interleukin-20 monoclonal antibody NNC0109-0012 in healthy volunteers and patients with psoriasis or rheumatoid arthritis. Adv Ther. 2015;32(3):228–38. 10.1007/s12325-015-0191-7
    1. World Medical Association. Declaration of Helsinki—Ethical Principles for Medical Research Involving Human Subjects 2004. Available: . Accessed: 10 February 2014.
    1. International Conference on Harmonisation. ICH Harmonised Tripartite Guideline. Good Clinical Practice. 01 May 1996.
    1. Robinson A, Kardos M, Kimball AB. Physician Global Assessment (PGA) and Psoriasis Area and Severity Index (PASI): why do both? A systematic analysis of randomized controlled trials of biologic agents for moderate to severe plaque psoriasis. J Am Acad Dermatol. 2012;66(3):369–75. 10.1016/j.jaad.2011.01.022
    1. Puzenat E, Bronsard V, Prey S, Gourraud PA, Aractingi S, Bagot M, et al. What are the best outcome measures for assessing plaque psoriasis severity? A systematic review of the literature. J Eur Acad Dermatol Venereol. 2010;24 Suppl 2:10–6. 10.1111/j.1468-3083.2009.03562.x
    1. Wang F, Smith N, Maier L, Xia W, Hammerberg C, Chubb H, et al. Etanercept suppresses regenerative hyperplasia in psoriasis by acutely downregulating epidermal expression of interleukin (IL)-19, IL-20 and IL-24. Br J Dermatol. 2012;167(1):92–102. 10.1111/j.1365-2133.2012.10961.x
    1. Garcia-Perez ME, Jean J, Pouliot R. Antipsoriatic drug development: challenges and new emerging therapies. Recent Pat Inflamm Allergy Drug Discov. 2012;6(1):3–21.
    1. Wang L, Li K, Xu Q, Chen X, Xue F, Shi R, et al. Potential synergy between SNP and CpG-A or IL-1beta in regulating transcriptional activity of IL-20 promoter. J Invest Dermatol. 2014;134(2):389–95. 10.1038/jid.2013.314

Source: PubMed

3
Suscribir