Pharmacodynamic Effects of Topical Omiganan in Patients With Mild to Moderate Atopic Dermatitis in a Randomized, Placebo-Controlled, Phase II Trial

Tessa Niemeyer-van der Kolk, Hein van der Wall, Geretta K Hogendoorn, Rianne Rijneveld, Sascha Luijten, Dirk C J G van Alewijk, Ellen H A van den Munckhof, Marieke L de Kam, Gary L Feiss, Errol P Prens, Jacobus Burggraaf, Robert Rissmann, Martijn B A van Doorn, Tessa Niemeyer-van der Kolk, Hein van der Wall, Geretta K Hogendoorn, Rianne Rijneveld, Sascha Luijten, Dirk C J G van Alewijk, Ellen H A van den Munckhof, Marieke L de Kam, Gary L Feiss, Errol P Prens, Jacobus Burggraaf, Robert Rissmann, Martijn B A van Doorn

Abstract

Omiganan is an indolicidin analog with antimicrobial properties that could be beneficial for patients with atopic dermatitis. In this randomized, double-blind, placebo-controlled, phase II trial we explored the efficacy, pharmacodynamics, and safety of topical omiganan once daily in 36 patients with mild to moderate atomic dermatitis. Patients were randomized to apply topical omiganan 1%, omiganan 2.5%, or vehicle gel to one target lesion once daily for 28 consecutive days. Small but significant improvements in local objective SCORing Atopic Dematitis index and morning itch were observed in the omiganan 2.5% group compared with the vehicle gel group (-18.5%; 95% confidence interval, -32.9 to -1.0; P = 0.04; and -8.2; 95% confidence interval, -16.3 to -0.2; P = 0.05, respectively). A shift from lesional to nonlesional skin microbiota was observed in both omiganan treatment groups, in contrast to the vehicle group. Thus, treatment with topical omiganan improved dysbiosis in patients with mild to moderate atopic dermatitis, and small but statistically significant improvements in clinical scores were detected. Our findings warrant further exploration in future clinical trials.

Trial registration: ClinicalTrials.gov NCT02456480.

Conflict of interest statement

G.L.F. was employed by Cutanea Life Sciences. The final draft was approved by G.L.F. on behalf of the sponsor, but no major comments were made. The authors declared no competing interests for this work.

© 2020 The Authors. Clinical and Translational Science published by Wiley Periodicals Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 2
Figure 2
Change from baseline in body surface area, objective SCORing Atopic Dematitis (oSCORAD) index and morning and evening itch in the omiganan 1% and 2.5% treatment groups compared with the vehicle gel group. Change from baseline graphs: delta least‐squares mean (LSM) data over time of clinical assessments for (a) body surface area (BSA) and oSCORAD (b) of the target lesion. In the lower panels, patient‐reported outcomes are depicted, i.e., itch in the (c) morning and (d) evening.
Figure 3
Figure 3
Pharmacodynamic effects of topical omiganan in the omiganan 1%, omiganan 2.5%, and vehicle gel groups. (a) Transepidermal water loss (TEWL) over time is depicted, showing improvement in the treatment groups and the vehicle group. Relative mRNA expressions in skin punch biopsy of markers (b) interleukin‐31, (c) eotaxin, and (d) interferon‐γ in lesional vs. nonlesional skin in mild to moderate atopic dermatitis patients at baseline. Data presented as median with interquartile range. Statistical significance is as follows: *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, based on a paired t‐test on log‐transformed data. (e) Relative mRNA expression of eotaxin per treatment group before treatment of lesional skin (day 0) and after end of treatment of lesional skin (day 28) and nonlesional (NL) skin. No treatment effect could be seen with this marker.
Figure 4
Figure 4
Course of cutaneous microbiome after omiganan treatment over time. Relative Staphylococcus abundance over time in the (a) omiganan 1%, (b) omiganan 2.5%, and (c) vehicle groups. A reduction is seen in both active treatment groups, but not in the vehicle group. Nonlesional boxes are presented as control on the right side. Median values are shown in blue above the boxplots.
Figure 5
Figure 5
Omiganan improves dysbiosis of the target lesion. (a) Staphylococcus abundance and (b) diversity index from baseline to day 28 (end of treatment) per treatment group, with P‐values of the differences calculated using a mixed model for data over time.
Figure 6
Figure 6
Correlation analysis of target lesion objective SCORing Atopic Dematitis (oSCORAD) index and Staphylococcus abundance. Correlations are shown between the (a) local target lesion oSCORAD index and Staphylococcus abundance in the microbiome and for the (b) target lesion oSCORAD and diversity index. (c,d) Delta correlations.
Figure 1
Figure 1
Flowchart of the study. QD = qualified dose (once daily).

References

    1. DaVeiga, S.P. Epidemiology of atopic dermatitis: a review. Allergy Asthma Proc. 33, 227–234 (2012).
    1. Bieber, T. Atopic dermatitis. N. Engl. J. Med. 358, 1483–1494 (2008).
    1. Tuffs, S.W. , Haeryfar, S.M.M. & McCormick, J.K. Manipulation of innate and adaptive immunity by staphylococcal superantigens. Pathogens. 7, 53 (2018).
    1. Park, H.Y. et al. Staphylococcus aureus colonization in acute and chronic skin lesions of patients with atopic dermatitis. Ann. Dermatol. 25, 410–416 (2013).
    1. Ong, P.Y. et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N. Engl. J. Med. 347, 1151–1160 (2002).
    1. Nomura, I. et al. Cytokine milieu of atopic dermatitis, as compared to psoriasis, skin prevents induction of innate immune response genes. J. Immunol. 171, 3262–3269 (2003).
    1. Kisich, K.O. , Carspecken, C.W. , Fieve, S. , Boguniewicz, M. & Leung, D.Y. Defective killing of Staphylococcus aureus in atopic dermatitis is associated with reduced mobilization of human beta‐defensin‐3. J. Allergy Clin. Immunol. 122, 62–68 (2008).
    1. Fritsche, T.R. , Rhomberg, P.R. , Sader, H.S. & Jones, R.N. In vitro activity of omiganan pentahydrochloride tested against vancomycin‐tolerant, ‐intermediate, and ‐resistant Staphylococcus aureus . Diagn. Microbiol. Infect. Dis. 60, 399–403 (2008).
    1. Fritsche, T.R. , Rhomberg, P.R. , Sader, H.S. & Jones, R.N. Antimicrobial activity of omiganan pentahydrochloride against contemporary fungal pathogens responsible for catheter‐associated infections. Antimicrob. Agents Chemother. 52, 1187–1189 (2008).
    1. Fritsche, T.R. , Rhomberg, P.R. , Sader, H.S. & Jones, R.N. Antimicrobial activity of omiganan pentahydrochloride tested against contemporary bacterial pathogens commonly responsible for catheter‐associated infections. J. Antimicrob. Chemother. 61, 1092–1098 (2008).
    1. Niyonsaba, F. , Kiatsurayanon, C. , Chieosilapatham, P. & Ogawa, H. Friends or foes? Host defense (antimicrobial) peptides and proteins in human skin diseases. Exp. Dermatol. 26, 989–998 (2017).
    1. van der Kolk, T. et al. Comprehensive, multimodal characterization of an imiquimod‐induced human skin inflammation model for drug development. Clin. Transl. Sci. 11, 607–615 (2018).
    1. Jensen, M.P. , Karoly, P. & Braver, S. The measurement of clinical pain intensity: a comparison of six methods. Pain 27, 117–126 (1986).
    1. Elman, S. , Hynan, L.S. , Gabriel, V. & Mayo, M.J. The 5‐D itch scale: a new measure of pruritus. Br. J. Dermatol. 162, 587–593 (2010).
    1. van den Munckhof, E.H.A. et al. Inter‐ and intra‐patient variability over time of lesional skin microbiota in adult patients with atopic dermatitis. Acta Derm. Venereol. 100, adv00018 (2020).
    1. Pichon, B. et al. Development of a real‐time quadruplex PCR assay for simultaneous detection of nuc, Panton‐Valentine leucocidin (PVL), mecA and homologue mecALGA251. J. Antimicrob. Chemother. 67, 2338–2341 (2012).
    1. Klindworth, A. et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next‐generation sequencing‐based diversity studies. Nucleic Acids Res. 41, e1 (2013).
    1. Niemeyer‐van der Kolk, T.P. et al. Omiganan enhances imiquimod‐induced inflammatory responses in a human skin challenge model. 2020;Feb 10. doi: 10.1111/cts.12741.
    1. Rijsbergen, M. et al. Mobile e‐diary application facilitates the monitoring of patient‐reported outcomes and a high treatment adherence for clinical trials in dermatology. J. Eur. Acad. Dermatol. Venereol. 34, 633–639 (2020).
    1. Kong, H.H. et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 22, 850–859 (2012).
    1. Kim, M.H. et al. A metagenomic analysis provides a culture‐independent pathogen detection for atopic dermatitis. Allergy Asthma Immunol. Res. 9, 453–461 (2017).
    1. Gonzalez, M.E. et al. Cutaneous microbiome effects of fluticasone propionate cream and adjunctive bleach baths in childhood atopic dermatitis. J. Am. Acad. Dermatol. 75, 481–493 (2016).
    1. Human Microbiome Project Consortium . A framework for human microbiome research. Nature 486, 215–221 (2012).
    1. Grice, E.A. & Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 9, 244–253 (2011).
    1. Bath‐Hextall, F.J. , Birnie, A.J. , Ravenscroft, J.C. & Williams, H.C. Interventions to reduce Staphylococcus aureus in the management of atopic eczema: an updated Cochrane review. Br. J. Dermatol. 163, 12–26 (2010).
    1. Dombrowski, Y. et al Cytosolic DNA triggers inflammasome activation in keratinocytes in psoriatic lesions. Sci. Translat. Med. 3, 82ra38 (2011).
    1. Kahlenberg, J.M. & Kaplan, M.J. Little peptide, big effects: the role of LL‐37 in inflammation and autoimmune disease. J. Immunol. 191, 4895–4901 (2013).
    1. Umehara, Y. , Kamata, Y. , Tominaga, M. , Niyonsaba, F. , Ogawa, H. & Takamori, K. Cathelicidin LL‐37 induces semaphorin 3A expression in human epidermal keratinocytes: implications for possible application to pruritus. J. Invest. Dermatol. 135, 2887–2890 (2015).
    1. Niyonsaba, F. et al. Antimicrobial peptides human beta‐defensins and cathelicidin LL‐37 induce the secretion of a pruritogenic cytokine IL‐31 by human mast cells. J. Immunol. 184, 3526–3534 (2010).
    1. Niyonsaba, F. , Someya, A. , Hirata, M. , Ogawa, H. & Nagaoka, I. Evaluation of the effects of peptide antibiotics human beta‐defensins‐1/‐2 and LL‐37 on histamine release and prostaglandin D(2) production from mast cells. Eur. J. Immunol. 31, 1066–1075 (2001).
    1. Niemeyer‐van der Kolk, T. , van der Wall, H.E.C. , Balmforth, C. , Van Doorn, M.B.A. & Rissmann, R. A systematic literature review of the human skin microbiome as biomarker for dermatological drug development. Br. J. Clin. Pharmacol. 84, 2178–2193 (2018).
    1. Copay, A.G. , Subach, B.R. , Glassman, S.D. , Polly, D.W. Jr. & Schuler, T.C. Understanding the minimum clinically important difference: a review of concepts and methods. Spine J. 7, 541–546 (2007).
    1. Reich, A. et al. Itch assessment with visual analogue scale and numerical rating scale: determination of minimal clinically important difference in chronic itch. Acta Dermato‐Venereologica. 96, 978–980 (2016).
    1. Lee, C.H. & Yu, H.S. Biomarkers for itch and disease severity in atopic dermatitis. Curr. Probl. Dermatol. 41, 136–148 (2011).

Source: PubMed

3
S'abonner