The effects of lebrikizumab in patients with mild asthma following whole lung allergen challenge

H Scheerens, J R Arron, Y Zheng, W S Putnam, R W Erickson, D F Choy, J M Harris, J Lee, N N Jarjour, J G Matthews, H Scheerens, J R Arron, Y Zheng, W S Putnam, R W Erickson, D F Choy, J M Harris, J Lee, N N Jarjour, J G Matthews

Abstract

Background: Interleukin 13 (IL13) is a T-helper type 2 (Th2) cytokine associated with inflammation and pathology in allergic diseases such as bronchial asthma. We have shown that treatment with lebrikizumab, an anti-IL13 monoclonal antibody, significantly improves prebronchodilator forced expiratory volume in 1 s (FEV(1)) in a subset of subjects with uncontrolled asthma.

Objective: To evaluate efficacy and safety of lebrikizumab in subjects with mild asthma who underwent bronchial allergen challenge.

Methods: Twenty-nine subjects were randomized 1 : 1-5 mg/kg lebrikizumab (n = 13) or placebo (n = 16) administered subcutaneously every 4 weeks over 12 weeks, a total of four doses. Primary efficacy outcome was late asthmatic response (LAR) at Week 13, defined as area under the curve of FEV1 measured 2-8 h following inhaled allergen challenge. Serum biomarkers were measured to verify IL13 pathway inhibition and identify patients with an increased response to lebrikizumab.

Results: At Week 13, the LAR in lebrikizumab subjects was reduced by 48% compared with placebo subjects, although this was not statistically significant (95% confidence interval, -19%, 90%). Exploratory analysis indicated that lebrikizumab-treated subjects with elevated baseline levels of peripheral blood eosinophils, serum IgE, or periostin exhibited a greater reduction in LAR compared with subjects with lower baseline levels of these biomarkers. Lebrikizumab exerted systemic effects on markers of Th2 inflammation, reducing serum immunoglobulin E (IgE), chemokine ligands 13 and 17 by approximately 25% (P < 0.01). Lebrikizumab was well tolerated.

Conclusion and clinical relevance: Lebrikizumab reduced the LAR in subjects with mild asthma. Clinical trial number NCT00781443.

Keywords: IL13; Th2 inflammation; allergen challenge; asthma; biomarkers; lebrikizumab.

© 2013 John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Study design. The study included a 21-day screening period, a 12-week treatment period, and a 16-week follow-up period. At screening and at Week 13, subjects underwent an allergen challenge followed within 18–24 h by a methacholine challenge to determine PC20, defined as the provocative concentration of methacholine that resulted in a > 20% reduction in FEV1. Subjects were randomized 1: 1 to lebrikizumab (5 mg/kg) or placebo, administered subcutaneously once every 4 weeks.
Figure 2
Figure 2
Allergen-induced changes in FEV1 following challenges at screening and Week 13. The allergen-induced percentage change in FEV1 during screening (a) and at Week 13 (b) in placebo- and lebrikizumab-treated subjects. FEV1 was collected every 10 min for the first 90 min and then every hour from 2 to 8 h following allergen challenge. Data points are means. Vertical bars are standard error of the mean.
Figure 3
Figure 3
Lebrikizumab appeared to have a greater but not statistically significant effect on allergen-induced late asthmatic response (LAR) in subjects with increased baseline levels of periostin, IgE, and/or peripheral blood eosinophils based on exploratory analysis. Black bars represent subjects with baseline levels of IgE, peripheral blood eosinophils, and periostin levels below the median of all subjects. Grey bars represent subjects with baseline levels of those biomarkers above the median of all subjects. Data are expressed as placebo-corrected mean LAR reduction at Week 13. The total number of subjects in active subgroups was low (5–8 active subjects/group), and statistical analyses were not performed on these exploratory endpoints.

References

    1. Saha SK, Berry MA, Parker D, et al. Increased sputum and bronchial biopsy IL-13 expression in severe asthma. J Allergy Clin Immunol. 2008;121:685–91.
    1. Humbert M, Durham SR, Kimmitt P, et al. Elevated expression of messenger ribonucleic acid encoding IL-13 in the bronchial mucosa of atopic and nonatopic subjects with asthma. J Allergy Clin Immunol. 1997;99:657–65.
    1. Truyen E, Coteur L, Dilissen E, et al. Evaluation of airway inflammation by quantitative Th1/Th2 cytokine mRNA measurement in sputum of asthma patients. Thorax. 2006;61:202–8.
    1. Kroegel C, Julius P, Matthys H, Virchow JC, Jr, Luttman W. Endobronchial secretion of interleukin-13 following local allergen challenge in atopic asthma: relationship to interleukin-4 and eosinophil counts. Eur Respir J. 1996;9:899–904.
    1. Huang SX, Xiao HQ, Kleine-Tebbe J, et al. IL-13 expression at the sites of allergen challenge in patients with asthma. J Immunol. 1995;155:2688–94.
    1. Ultsch M, Bevers J, Nakamura G, et al. Structural basis of signaling blockade by anti-IL-13 antibody lebrikizumab. J Mol Biol. 2013;425:1330–9.
    1. Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med. 2011;365:1088–98.
    1. O’Byrne PM, Gauvreau GM, Brannan JD. Provoked models of asthma: what have we learnt? Clin Exp Allergy. 2009;39:181–92.
    1. Wenzel S, Wilbraham D, Fuller R, Getz EB, Longphre M. Effect of an interleukin-4 variant on late phase asthmatic response to allergen challenge in asthmatic patients: results of two phase 2a studies. Lancet. 2007;370:1422–31.
    1. Gauvreau GM, Boulet LP, Cockcroft DW, et al. The effects of interleukin-13 blockade on allergen-induced airway responses in mild atopic asthma. Am J Respir Crit Care Med. 2011;183:1007–14.
    1. Slager RE, Hawkins GA, Ampleford EJ, et al. IL-4 receptor alpha polymorphisms are predictors of a pharmacogenetic response to a novel IL-4/IL-13 antagonist. J Allergy Clin Immunol. 2010;126:875–8.
    1. Haldar P, Pavord ID. Noneosinophilic asthma: a distinct clinical and pathologic phenotype. J Allergy Clin Immunol. 2007;119:1043–52.
    1. Anderson GP. Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet. 2008;372:1107–19.
    1. Moore WC, Meyers DA, Wenzel SE, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med. 2010;181:315–23.
    1. Simpson JL, Scott R, Boyle MJ, Gibson PG. Inflammatory subtypes in asthma: assessment and identification using induced sputum. Respirology. 2006;11:54–61.
    1. Wardlaw AJ, Silverman M, Siva R, Pavord ID, Green R. Multi-dimensional phenotyping: towards a new taxonomy for airway disease. Clin Exp Allergy. 2005;35:1254–62.
    1. Jia G, Erickson R, Choy D, et al. Periostin is a systemic biomarker of eosinophilic airway inflammation in asthmatic patients.; Bronchoscopic Exploratory Research Study of Biomarkers in Corticosteroid-refractory Asthma (BOBCAT) Study Group. J Allergy Clin Immunol. 2012;130:647–54.
    1. Corren J, Busse W, Meltzer EO, Mansfield L, Bensch G, Fahrenholz J. A randomized, controlled, phase 2 study of AMG 317, an IL-4Ralpha antagonist, in patients with asthma. Am J Respir Crit Care Med. 2010;181:788–96.
    1. Woodruff PG, Modrek B, Choy DF, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009;180:388–95.
    1. Choy DF, Modrek B, Abbas AR, et al. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. J Immunol. 2011;186:1861–9.
    1. Sekiya T, Yamada H, Yamaguchi M, et al. Increased levels of a TH2-type CC chemokine thymus and activation-regulated chemokine (TARC) in serum and induced sputum of asthmatics. Allergy. 2002;57:173–7.
    1. Chupp GL, Lee CG, Jarjour N, et al. A chitinase-like protein in the lung and circulation of patients with severe asthma. N Engl J Med. 2007;357:2016–27.
    1. Maeda Y, Hizawa N, Fukui Y, et al. Concentrations of carcinoembryonic antigen in serum and bronchoalveolar lavage fluid of asthmatic patients with mucoid impaction. Nihon Kokyuki Gakkai Zasshi. 2004;42:988–93.
    1. Dougherty RH, Sidhu SS, Raman K, et al. Accumulation of intraepithelial mast cells with a unique protease phenotype in T(H)2-high asthma. J Allergy Clin Immunol. 2010;125:1046–53.
    1. Crapo RO, Casaburi R, Coates AL, et al. Guidelines for methacholine and exercise challenge testing – 1999: the official statement of the American Thoracic Society. Am J Respir Crit Care Med. 2000;161:309–29.
    1. Crapo RO. Spirometry: quality control and reproducibility criteria. Am Rev Respir Dis. 1991;143:1212–3.
    1. Juniper EF, Cockroft DW, Hargreave FE. Histamine and methacholine inhalation tests; a laboratory tidal breathing tidal protocol. Lund, Sweden: Astra Draco AB; 1994.
    1. Fahy JV, Fleming HE, Wong HH, et al. The effect of an anti-IgE monoclonal antibody on the early- and late-phase responses to allergen inhalation in asthmatic subjects. Am J Respir Crit Care Med. 1997;155:1828–34.
    1. van Rensen EL, Evertse CE, van Schadewijk WA, et al. Eosinophils in bronchial mucosa of asthmatics after allergen challenge: effect of anti-IgE treatment. Allergy. 2009;64:72–80.
    1. Leckie MJ, ten Brinke A, Khan J, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356:2144–8.
    1. Flood-Page PT, Menzies-Gow AN, Kay AB, Robinson DS. Eosinophil’s role remains uncertain as anti-interleukin-5 only partially depletes numbers in asthmatic airway. Am J Crit Care Med. 2003;167:199–204.
    1. Gauvreau GM, Watson RM, Rerecich TJ, Baswick E, Inman MD, O’Byrne PM. Repeatability of allergen-induced airway inflammation. J Allergy Clin Immunol. 1999;104:66–71.
    1. Kalayci O, Sonna LA, Woodruff PG, Camargo CA, Jr, Luster AD, Lilly CM. Monocyte chemotactic protein-4 (MCP-4; CCL-13): a biomarker of asthma. J Asthma. 2004;41:27–33.
    1. Leung TF, Wong CK, Chan IH, Ip WK, Lam CW, Wong GW. Plasma concentration of thymus and activation-regulated chemokine is elevated in childhood asthma. J Allergy Clin Immunol. 2002;110:404–9.

Source: PubMed

3
Iratkozz fel