Safety, pharmacokinetics and pharmacodynamics of a topical SYK inhibitor in cutaneous lupus erythematosus: A double-blind Phase Ib study

Alex Walker, Lars Erwig, Katie Foster, Katherine Nevin, Joerg Wenzel, Margitta Worm, Nicola Williams, Nirav Ratia, Bao Hoang, Tanja Schneider-Merck, Sophie Gisbert, Heike Carnarius, Marion Dickson, Alex Walker, Lars Erwig, Katie Foster, Katherine Nevin, Joerg Wenzel, Margitta Worm, Nicola Williams, Nirav Ratia, Bao Hoang, Tanja Schneider-Merck, Sophie Gisbert, Heike Carnarius, Marion Dickson

Abstract

The immunoregulator spleen tyrosine kinase (SYK) is upregulated in cutaneous lupus erythematosus (CLE). This double-blind, multicentre, Phase Ib study evaluated the safety, tolerability, pharmacokinetics, pharmacodynamics and clinical efficacy of the selective SYK inhibitor GSK2646264 in active CLE lesions. Two lesions from each participant (n = 11) were each randomized to topical application of 1% (w/w) GSK2646264 or placebo for 28 days; all participants received GSK2646264 and placebo. The primary endpoint was safety and tolerability of GSK2646264, assessed by adverse event incidence and a skin tolerability test. Secondary endpoints included change from baseline in clinical activity and mRNA expression of interferon-related genes in skin biopsies. Levels of several immune cell markers were evaluated over time. Eight (73%) participants experienced ≥ 1 adverse event (all mild in intensity), and maximal dermal response was similar for GSK2646264 and placebo. The expression of several interferon-related genes, including CXCL10 and OAS1, showed modest decreases from baseline after 28 days of treatment with GSK2646264 compared with placebo. Similar findings were observed for CD3 + T cell and CD11c + dendritic cell levels; however, overall clinical activity remained unchanged with GSK2646264 vs. placebo. Further studies are warranted to assess SYK inhibitors as potential treatment for CLE.

Trial registration: ClinicalTrials.gov NCT02927457.

Keywords: SYK Kinase; cutaneous lupus erythematosus; interferons; pharmacology; safety.

Conflict of interest statement

AW, KF, KN, NW, NR, BH, TSM, SG and HC are employees of GSK and hold stocks/shares in GSK. LE and MD were employees of GSK at the time of the study. JW reports grants from GSK, grants from Incyte, personal fees from Biogen, personal fees from Leo Pharma and other from Novartis. MW received honoraria for consulting by ALK‐Abelló Arzneimittel GmbH, Mylan Germany GmbH, Bencard Allergie GmbH, Novartis AG, Biotest AG, Actelion Pharmaceuticals Deutschland GmbH, Sanofi‐Aventis Deutschland GmbH and HAL Allergie GmbH.

© 2020 Glaxo Group Limited. Experimental Dermatology published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Adjusted mean (95% CI) intensity of log2 mRNA expression by visit and treatment (A) CXCL10, (B) IFI44, (C) IFIH1, (D) OAS1, (E) IL1A, (F) IL1B, and (G) IL6, (H) IFI16, (I) IFI44L, (J) IFIT1 and (K) IFIT3. For genes that had more than one probe analysed, the probe that showed the largest treatment difference is shown. Adjusted mean intensity values were derived using a mixed model with participant as a random effect and treatment as a fixed effect where treatment is set to “not applicable” at baseline. CI, confidence interval
FIGURE 2
FIGURE 2
Mean (±SE) expression (cells/mm2) of (A) CD3 + T cells and (B) CD11c + dendritic cells in the dermis from biopsies by visit, treatment and sub‐acute/chronic CLE subtypes. Immunohistochemical staining (C) of CD3 + T cells (i–v, xi–xv) and CD11C + dendritic cells (vi–x, xvi–xx) in CLE skin samples in the presence and absence of GSK2646264 treatment from two representative participants. Images represent uninvolved (i, vi, xi, xvi), baseline placebo (ii, vii, xii, xvii), baseline GSK2646264 (iii, viii, xiii, xviii), placebo‐treated at Day 28 (iv, ix, ixv, ixx) and GSK2646264‐treated (x, v, xv, xx) at Day 28. Yellow staining = CD3/CD11c; blue staining = haematoxylin; green = overlap of yellow and blue staining. Scale bars denote 200 µm. Images from Participant #8 and Participant #9 are included as representative images for participants in which immunohistochemical staining changes were and were not observed, respectively. CLE, chronic lupus erythematosus; SE, standard error

References

    1. Okon LG, Werth VP. Cutaneous lupus erythematosus: diagnosis and treatment. Best Pract Res Clin Rheumatol. 2013;27:391‐404. 10.1016/j.berh.2013.07.008
    1. Presto JK, Hejazi EZ, Werth VP. Biological therapies in the treatment of cutaneous lupus erythematosus. Lupus. 2017;26:115‐118. 10.1177/0961203316670731
    1. Ogunsanya ME, Brown CM, Lin D, Imarhia F, Maxey C, Chong BF. Understanding the disease burden and unmet needs among patients with cutaneous lupus erythematosus: A qualitative study. Int J Womens Dermatol. 2018;4:152‐158. 10.1016/j.ijwd.2018.01.002
    1. Hall SA, Chen SY, Kao A, et al. 162 Symptom impact and unmet need in systemic/cutaneous lupus erythematosus: results from a patient‐centred study set in a social media community. Lupus Sci Med. 2017;4:A77. 10.1136/lupus-2017-000215.162
    1. Werth V, Eldar‐Lissai A, Ning W. 155 Disease burden and healthcare resource utilisation among cutaneous lupus erythematosus patients with depression and/or anxiety; quantifying the unmet need. Lupus Sci Med. 2017;4:A71. 10.1136/lupus-2017-000215.155
    1. Braegelmann C, Holzel M, Ludbrook V, et al. Spleen tyrosine kinase (SYK) is a potential target for the treatment of cutaneous lupus erythematosus patients. Exp Dermatol. 2016;25:375‐379. 10.1111/exd.12986
    1. Mócsai A, Ruland J, Tybulewicz VLJ. The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol. 2010;10:387‐402. 10.1038/nri2765
    1. Barker MD, Liddle J, Atkinson FL, et al. Discovery of potent and selective Spleen Tyrosine Kinase inhibitors for the topical treatment of inflammatory skin disease. Bioorg Med Chem Lett. 2018;28:3458‐3462. 10.1016/j.bmcl.2018.09.022
    1. Ramirez Molina C, Falkencrone S, Skov PS, Hooper‐Greenhill E, Barker M, Dickson MC. GSK2646264, a spleen tyrosine kinase inhibitor, attenuates the release of histamine in ex vivo human skin. Br J Pharmacol. 2019;176:1135‐1142. 10.1111/bph.14610
    1. Kuhn A, Meuth AM, Bein D, et al. Revised Cutaneous Lupus Erythematosus Disease Area and Severity Index (RCLASI): a modified outcome instrument for cutaneous lupus erythematosus. Br J Dermatol. 2010;163:83‐92. 10.1111/j.1365-2133.2010.09799.x
    1. Li H, Ice JA, Lessard CJ, Sivils KL. Interferons in Sjögren's syndrome: genes, mechanisms, and effects. Front Immunol. 2013;4:290. 10.3389/fimmu.2013.00290
    1. Braunstein I, Klein R, Okawa J, Werth VP. The interferon‐regulated gene signature is elevated in subacute cutaneous lupus erythematosus and discoid lupus erythematosus and correlates with the cutaneous lupus area and severity index score. Br J Dermatol. 2012;166:971‐975. 10.1111/j.1365-2133.2012.10825.x
    1. Chuang JY, Huang YL, Yen WL, Chiang IP, Tsai MH, Tang CH. Syk/JNK/AP‐1 signaling pathway mediates interleukin‐6‐promoted cell migration in oral squamous cell carcinoma. Int J Mol Sci. 2014;15:545‐559. 10.3390/ijms15010545
    1. Deng GM, Liu L, Bahjat FR, Pine PR, Tsokos GC. Suppression of skin and kidney disease by inhibition of spleen tyrosine kinase in lupus‐prone mice. Arthritis Rheum. 2010;62:2086‐2092. 10.1002/art.27452
    1. Guiducci C, Tripodo C, Gong M, et al. Autoimmune skin inflammation is dependent on plasmacytoid dendritic cell activation by nucleic acids via TLR7 and TLR9. J Exp Med. 2010;207:2931‐2942. 10.1084/jem.20101048
    1. Hussein MR, Aboulhagag NM, Atta HS, Atta SM. Evaluation of the profile of the immune cell infiltrate in lichen planus, discoid lupus erythematosus, and chronic dermatitis. Pathology. 2008;40:682‐693. 10.1080/00313020802320739
    1. Chen IH, Xue L, Hsu CC, et al. Phosphoproteins in extracellular vesicles as candidate markers for breast cancer. Proc Natl Acad Sci USA. 2017;114:3175‐3180. 10.1073/pnas.1618088114

Source: PubMed

3
Suscribir