A randomized, placebo-controlled trial evaluating effects of lebrikizumab on airway eosinophilic inflammation and remodelling in uncontrolled asthma (CLAVIER)

Cary D Austin, Melissa Gonzalez Edick, Ronald E Ferrando, Margaret Solon, Miriam Baca, Kathryn Mesh, Peter Bradding, Gail M Gauvreau, Kaharu Sumino, J Mark FitzGerald, Elliot Israel, Lief Bjermer, Arnaud Bourdin, Joseph R Arron, David F Choy, Julie K Olsson, Francis Abreu, Monet Howard, Kit Wong, Fang Cai, Kun Peng, Wendy S Putnam, Cécile T J Holweg, John G Matthews, Monica Kraft, Prescott G Woodruff, CLAVIER Investigators, Cary D Austin, Melissa Gonzalez Edick, Ronald E Ferrando, Margaret Solon, Miriam Baca, Kathryn Mesh, Peter Bradding, Gail M Gauvreau, Kaharu Sumino, J Mark FitzGerald, Elliot Israel, Lief Bjermer, Arnaud Bourdin, Joseph R Arron, David F Choy, Julie K Olsson, Francis Abreu, Monet Howard, Kit Wong, Fang Cai, Kun Peng, Wendy S Putnam, Cécile T J Holweg, John G Matthews, Monica Kraft, Prescott G Woodruff, CLAVIER Investigators

Abstract

Background: The anti-interleukin 13 (IL-13) monoclonal antibody lebrikizumab improves lung function in patients with moderate-to-severe uncontrolled asthma, but its effects on airway inflammation and remodelling are unknown. CLAVIER was designed to assess lebrikizumab's effect on eosinophilic inflammation and remodelling.

Objective: To report safety and efficacy results from enrolled participants with available data from CLAVIER.

Methods: We performed bronchoscopy on patients with uncontrolled asthma before and after 12 weeks of randomized double-blinded treatment with lebrikizumab (n = 31) or placebo (n = 33). The pre-specified primary end-point was relative change in airway subepithelial eosinophils per mm2 of basement membrane (cells/mm2 ). Pre-specified secondary and exploratory outcomes included change in IL-13-associated biomarkers and measures of airway remodelling.

Results: There was a baseline imbalance in tissue eosinophils and high variability between treatment groups. There was no discernible change in adjusted mean subepithelial eosinophils/mm2 in response to lebrikizumab (95% CI, -82.5%, 97.5%). As previously observed, FEV1 increased after lebrikizumab treatment. Moreover, subepithelial collagen thickness decreased 21.5% after lebrikizumab treatment (95% CI, -32.9%, -10.2%), and fractional exhaled nitric oxide, CCL26 and SERPINB2 mRNA expression in bronchial tissues also reduced. Lebrikizumab was well tolerated, with a safety profile consistent with other lebrikizumab asthma studies.

Conclusions & clinical relevance: We did not observe reduced tissue eosinophil numbers in association with lebrikizumab treatment. However, in pre-specified exploratory analyses, lebrikizumab treatment was associated with reduced degree of subepithelial fibrosis, a feature of airway remodelling, as well as improved lung function and reduced key pharmacodynamic biomarkers in bronchial tissues. These results reinforce the importance of IL-13 in airway pathobiology and suggest that neutralization of IL-13 may reduce asthmatic airway remodelling.

Clinical trial registration: NCT02099656.

Conflict of interest statement

All authors report support of the parent study and funding of editorial support from F. Hoffmann‐La Roche. CDA, MGE, RF, MS, MB, KM, JA, DC, JO, FA, KP, MH, KW, FC, WSP and CTJH are employees of Genentech, Inc. JGM was an employee at Genentech, Inc, at the time of the study but is now an employee of 23andMe. PB has received grant funding from Genentech, Inc. GMG has received research funding from Genentech, Inc, paid directly to McMaster University. MF has received research funding from Genentech, Inc, paid directly to UBC. EI has received grants and nonfinancial support from Genentech during the conduct of the study. MK receives research funding for asthma (paid to the University of Arizona) from the National Institutes of Health, American Lung Association, AstraZeneca and Sanofi. MK engages in consulting activities with AstraZeneca and Sanofi and receives royalties from Elsevier. AB has received personal fees and nonfinancial support from Roche outside the submitted work. PW reports fees to his institution for assistance with analysis of samples in this clinical study and personal fees from Amgen, NGM biopharmaceuticals, Theravance, Clarus Ventures, AstraZeneca, 23andMe, Sanofi, Regeneron and GSK outside the submitted work.

© 2020 The Authors. Clinical & Experimental Allergy published by John Wiley & Sons Ltd.

Figures

Figure 1
Figure 1
Trial design, with lebrikizumab 125 mg or placebo administered subcutaneously on day 1, day 8, week 4 and week 8 during the 12‐week placebo‐controlled period. SC, subcutaneous
Figure 2
Figure 2
Patient disposition
Figure 3
Figure 3
Mean relative (%) changes from baseline in number of airway subepithelial eosinophils per mm2 of basement membrane at week 12 in the primary analysis population. Estimates were based on a linear model that used relative change from baseline in airway subepithelial eosinophils as the response variable and included terms for treatment, number of asthma exacerbations within 12 mo of study entry and baseline asthma medications. Relative change was defined as the absolute change from baseline to week 12 divided by the value at baseline. Placebo‐corrected adjusted mean change is the difference in adjusted mean changes between the lebrikizumab and placebo groups. Boxplots of the corresponding unadjusted data are provided in Figure S2
Figure 4
Figure 4
Mean adjusted relative (%) changes from baseline in thickness of subepithelial collagen at week 12 in the primary analysis population. Estimates were based on a linear model that used relative change from baseline in thickness of subepithelial collagen as the response variable and included terms for treatment, number of asthma exacerbations within 12 months of study entry and baseline asthma medications. Relative change was defined as the absolute change from baseline to week 12 divided by the value at baseline. Placebo‐corrected adjusted mean change is the difference in adjusted mean changes between the lebrikizumab and placebo groups. Boxplots of the corresponding unadjusted data are provided in Figure S7

References

    1. Braman SS. The global burden of asthma. Chest. 2006;130(1 Suppl):4S‐12S.
    1. Wenzel SE. Asthma: defining of the persistent adult phenotypes. Lancet. 2006;368(9537):804‐813.
    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(4):315‐323.
    1. Global Initiative for Asthma . 2019 GINA Report, Global Strategy for Asthma Management and Prevention, 2019.
    1. Lemiere C, Ernst P, Olivenstein R, et al. Airway inflammation assessed by invasive and noninvasive means in severe asthma: eosinophilic and none‐osinophilic phenotypes. J Allergy Clin Immunol. 2006;118(5):1033‐1039.
    1. Arron JR, Choy DF, Laviolette M, et al. Disconnect between sputum neutrophils and other measures of airway inflammation in asthma. Eur Respir J. 2014;43(2):627‐629.
    1. Jia G, Erickson RW, Choy DF, et al. Periostin is a systemic biomarker of eosinophilic airway inflammation in asthmatic patients. J Allergy Clin Immunol. 2012;130(3):647‐654.e610.
    1. Nair P, Ochkur SI, Protheroe C, et al. Eosinophil peroxidase in sputum represents a unique biomarker of airway eosinophilia. Allergy. 2013;68(9):1177‐1184.
    1. de Kluijver J, Schrumpf JA, Evertse CE, et al. Bronchial matrix and inflammation respond to inhaled steroids despite ongoing allergen exposure in asthma. Clin Exp Allergy. 2005;35(10):1361‐1369.
    1. Hershey GK. IL‐13 receptors and signaling pathways: an evolving web. J Allergy Clin Immunol. 2003;111(4):677‐690; quiz 691.
    1. Horie S, Okubo Y, Hossain M, et al. Interleukin‐13 but not interleukin‐4 prolongs eosinophil survival and induces eosinophil chemotaxis. Int Med. 1997;36(3):179‐185.
    1. Luttmann W, Knoechel B, Foerster M, Matthys H, Virchow JC Jr, Kroegel C. Activation of human eosinophils by IL‐13. Induction of CD69 surface antigen, its relationship to messenger RNA expression, and promotion of cellular viability. J Immunol. 1996;157(4):1678‐1683.
    1. Pope SM, Brandt EB, Mishra A, et al. IL‐13 induces eosinophil recruitment into the lung by an IL‐5‐ and eotaxin‐dependent mechanism. J Allergy Clin Immunol. 2001;108(4):594‐601.
    1. Zhu Z, Homer RJ, Wang Z, et al. Pulmonary expression of interleukin‐13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J Clin Investig. 1999;103(6):779‐788.
    1. Kumar RK, Herbert C, Yang M, Koskinen AM, McKenzie AN, Foster PS. Role of interleukin‐13 in eosinophil accumulation and airway remodelling in a mouse model of chronic asthma. Clin Exper Allergy. 2002;32(7):1104‐1111.
    1. Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med. 2011;365(12):1088‐1098.
    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(8):1330‐1339.
    1. Hanania NA, Korenblat P, Chapman KR, et al. Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II): replicate, phase 3, randomised, double‐blind, placebo‐controlled trials. Lancet Respir Med. 2016;4(10):781‐796.
    1. Doran E, Cai F, Holweg CTJ, Wong K, Brumm J, Arron JR. Interleukin‐13 in asthma and other eosinophilic disorders. Front Med. 2017;4:139.
    1. Arron JR, Corren J, Matthews JG. Lebrikizumab treatment in adults with asthma [letter]. N Engl J Med. 2011;365(25):2432‐2434.
    1. Hanania NA, Noonan M, Corren J, et al. Lebrikizumab in moderate‐to‐severe asthma: pooled data from two randomised placebo‐controlled studies. Thorax. 2015;70(8):748‐756.
    1. Olaguibel J, Quirce S, Juliá B, et al. Measurement of asthma control according to Global Initiative for Asthma guidelines: a comparison with the Asthma Control Questionnaire. Respir Res. 2012;13(1):9913‐9950.
    1. Noonan M, Korenblat P, Mosesova S, et al. Dose‐ranging study of lebrikizumab in asthmatic patients not receiving inhaled steroids. J Allergy Clin Immunol. 2013;132(3):567‐574.e512.
    1. Djukanović R, Wilson SJ, Kraft M, et al. Effects of treatment with anti‐immunoglobulin E antibody omalizumab on airway inflammation in allergic asthma. Am J Respir Crit Care Med. 2004;170(6):583‐593.
    1. Menzies‐Gow A, Flood‐Page P, Sehmi R, et al. Anti‐IL‐5 (mepolizumab) therapy induces bone marrow eosinophil maturational arrest and decreases eosinophil progenitors in the bronchial mucosa of atopic asthmatics. J Allergy Clin Immunol. 2003;111(4):714‐719.
    1. Acharya KR, Ackerman SJ. Eosinophil granule proteins: form and function. J Biol Chem. 2014;289(25):17406‐17415.
    1. Ochkur SI, Kim JD, Protheroe CA, et al. A sensitive high throughput ELISA for human eosinophil peroxidase: a specific assay to quantify eosinophil degranulation from patient‐derived sources. J Immunol Methods. 2012;384(1–2):10‐20.
    1. Mesnil C, Raulier S, Paulissen G, et al. Lung‐resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Investig. 2016;126(9):3279‐3295.
    1. Chetta A, Foresi A, Del Donno M, Bertorelli G, Pesci A, Olivieri D. Airways remodeling is a distinctive feature of asthma and is related to severity of disease. Chest. 1997;111(4):852‐857.
    1. Lee CG, Homer RJ, Zhu Z, et al. Interleukin‐13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor β1 . J Exper Med. 2001;194(6):809‐821.
    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(5):388‐395.
    1. Hoshino M, Nakamura Y, Sim JJ, et al. Inhaled corticosteroid reduced lamina reticularis of the basement membrane by modulation of insulin‐like growth factor (IGF)‐I expression in bronchial asthma. Clin Exper Allergy. 1998;28(5):568‐577.
    1. Olivieri D, Chetta A, Del Donno M, et al. Effect of short‐term treatment with low‐dose inhaled fluticasone propionate on airway inflammation and remodeling in mild asthma: a placebo‐controlled study. Am J Respir Crit Care Med. 1997;155(6):1864‐1871.
    1. Russell RJ, Chachi L, FitzGerald JM, et al. Effect of tralokinumab, an interleukin‐13 neutralising monoclonal antibody, on eosinophilic airway inflammation in uncontrolled moderate‐to‐severe asthma (MESOS): a multicentre, double‐blind, randomised, placebo‐controlled phase 2 trial. Lancet Respir Med. 2018;6(7):499‐510.
    1. Nyenhuis SM, Krishnan JA, Berry A, et al. Race is associated with differences in airway inflammation in patients with asthma. J Allergy Clin Immunol. 2017;140(1):257‐265.e211.

Source: PubMed

3
Abonneren