Airway remodelling rather than cellular infiltration characterizes both type2 cytokine biomarker-high and -low severe asthma

Latifa Khalfaoui, Fiona A Symon, Simon Couillard, Beverley Hargadon, Rekha Chaudhuri, Steve Bicknell, Adel H Mansur, Rahul Shrimanker, Timothy S C Hinks, Ian D Pavord, Stephen J Fowler, Vanessa Brown, Lorcan P McGarvey, Liam G Heaney, Cary D Austin, Peter H Howarth, Joseph R Arron, David F Choy, Peter Bradding, Latifa Khalfaoui, Fiona A Symon, Simon Couillard, Beverley Hargadon, Rekha Chaudhuri, Steve Bicknell, Adel H Mansur, Rahul Shrimanker, Timothy S C Hinks, Ian D Pavord, Stephen J Fowler, Vanessa Brown, Lorcan P McGarvey, Liam G Heaney, Cary D Austin, Peter H Howarth, Joseph R Arron, David F Choy, Peter Bradding

Abstract

Background: The most recognizable phenotype of severe asthma comprises people who are blood eosinophil and FeNO-high, driven by type 2 (T2) cytokine biology, which responds to targeted biological therapies. However, in many people with severe asthma, these T2 biomarkers are suppressed but poorly controlled asthma persists. The mechanisms driving asthma in the absence of T2 biology are poorly understood.

Objectives: To explore airway pathology in T2 biomarker-high and -low severe asthma.

Methods: T2 biomarker-high severe asthma (T2-high, n = 17) was compared with biomarker-intermediate (T2-intermediate, n = 21) and biomarker-low (T2-low, n = 20) severe asthma and healthy controls (n = 28). Bronchoscopy samples were processed for immunohistochemistry, and sputum for cytokines, PGD2 and LTE4 measurements.

Results: Tissue eosinophil, neutrophil and mast cell counts were similar across severe asthma phenotypes and not increased when compared to healthy controls. In contrast, the remodelling features of airway smooth muscle mass and MUC5AC expression were increased in all asthma groups compared with health, but similar across asthma subgroups. Submucosal glands were increased in T2-intermediate and T2-low asthma. In spite of similar tissue cellular inflammation, sputum IL-4, IL-5 and CCL26 were increased in T2-high versus T2-low asthma, and several further T2-associated cytokines, PGD2 and LTE4 , were increased in T2-high and T2-intermediate asthma compared with healthy controls.

Conclusions: Eosinophilic tissue inflammation within proximal airways is suppressed in T2 biomarker-high and T2-low severe asthma, but inflammatory and structural cell activation is present, with sputum T2-associated cytokines highest in T2 biomarker-high patients. Airway remodelling persists and may be important for residual disease expression beyond eosinophilic exacerbations. Registered at ClincialTrials.gov: NCT02883530.

Keywords: FeNO; Th2; cytokine; eosinophil; severe asthma.

Conflict of interest statement

SC has received non‐restricted research grants from the NIHR Oxford BRC, Sanofi‐Genzyme and the Quebec Respiratory Health Research Network; he is the holder of the Association Pulmonaire du Québec's Research Chair in Respiratory medicine; he received speaker honoraria from AstraZeneca, GlaxoSmithKline, Sanofi‐Regeneron and Valeo Pharma; he received consultancy fees for FirstThought; he has received sponsorship to attend international scientific meetings by AstraZeneca. He is an advisory board member for Biometry Inc—a company which is developing a FeNO device (myBiometry); the contract will be remunerated by stock options. RC has received lecture fees from GSK, AstraZeneca, Teva, Chiesi and Sanofi; honoraria for Advisory Board Meetings from GSK, AstraZeneca, Teva, Chiesi and Novarti; sponsorship to attend international scientific meetings from Chiesi, Napp, Sanofi, Boehringer, GSK and AstraZeneca and a research grant to her Institute from AstraZeneca for a UK multi‐centre study. AHM has received personal and institutional payment for talks, advisory board meetings, education and research funding from GSK, AstraZeneca, Teva, Chiesi, NAPP, Sanofi, Novartis, PI. TSCH has received grants from Pfizer Inc., the University of Oxford, the Wellcome Trust, The Guardians of the Beit Fellowship, the NIHR Oxford Biomedical Research Centre, Sensyne Health and Kymab during the conduct of the study; and personal fees from AstraZeneca, TEVA and Peer Voice outside the submitted work. IDP, In the last 5 years, IDP has received speaker's honoraria for speaking at sponsored meetings from Astra Zeneca, Boehringer Ingelheim, Aerocrine, Almirall, Novartis, Teva, Chiesi, Sanofi/Regeneron, Menarini and GSK, and payments for organizing educational events from AstraZeneca, GSK, Sanofi/Regeneron and Teva. He has received honoraria for attending advisory panels with Genentech, Sanofi/Regeneron, Astra Zeneca, Boehringer Ingelheim, GSK, Novartis, Teva, Merck, Circassia, Chiesi and Knopp, and payments to support FDA approval meetings from GSK. He has received sponsorship to attend international scientific meetings from Boehringer Ingelheim, GSK, AstraZeneca, Teva and Chiesi. He has received a grant from Chiesi to support a phase 2 clinical trial in Oxford. In 2014–5 and 2019–20, he was an expert witness for a patent dispute involving AstraZeneca and Teva. SJF has received grants from Boehringer Ingelheim and fees from AstraZeneca, Boehringer Ingelheim, Novartis, Teva and Chiesi. LPM declares Research funding from Chiesi and Merck; consultancies for Chiesi, Glaxo Smith Kline, Merck, Sanofi, Genentech and support to attend scientific meetings from Chiesi, Merck and Bayer. LGH reports grants from Genentech/Hoffman la Roche, during the conduct of the study; other from AstraZeneca, Boehringer Ingelheim, Chiesi, GSK and Napp Pharmaceuticals, personal fees from Novartis, Hoffman la Roche/Genentech Inc, Sanofi, Evelo Biosciences, Glaxo Smith Kline, Astra Zeneca, Teva, Theravance, Circassia, grants from Medimmune, Novartis UK, Roche/Genentech Inc and Glaxo Smith Kline, Amgen, Genentech/Hoffman la Roche, Astra Zeneca, Medimmune, Glaxo Smith Kline, Aerocrine and Vitalograph, outside the submitted work. CDA is an employee of Genentech. PHH is an employee of GSK. JRA is an employee of Genentech. DC is an employee of Genentech. PB has received research funding from Genentech via the University Hospitals of Leicester NHS Trust; consultancies for Boehringer Ingelheim, Genentech and Celldex Therapeutics via the University of Leicester. Support to attend scientific meetings from Chiesi, Teva and Sanofi‐Genzyme. BH, LC, FAS, SB, RS, VB no conflict of interest.

© 2022 The Authors. Allergy published by European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Eosinophil and neutrophil numbers in asthma subgroups and healthy controls. A) Eosinophil numbers in the airway lamina propria (Kruskal–Wallis p = 0.8444). B) Neutrophil numbers in the airway lamina propria (Kruskal–Wallis p = 0.0119). C) Neutrophil numbers in the airway epithelium (Kruskal–Wallis: p = 0.0197 all groups, p = 0.0105 asthma groups). **p = 0.0043 compared with healthy controls, #p = 0.0120 compared with T2‐intermediate (Dunn's multiple comparison test). Red points represent people taking oral corticosteroids
FIGURE 2
FIGURE 2
Mast cell numbers in asthma subgroups and healthy controls. A) Tryptase+ mast cell numbers in the airway lamina propria (ANOVA p < 0.0001). B) Chymase+ mast cell numbers in the airway lamina propria (Kruskal–Wallis p < 0.0001). C) Tryptase+ mast cell numbers within the airway smooth muscle (ASM) (Kruskal Wallis p = 0.0923). D) Chymase+ mast cell numbers within the ASM (Kruskal–Wallis p = 0.1346). E) The chymase:tryptase ratio in the lamina propria (Kruskal–Wallis p = 0.0280). F) The chymase:tryptase ratio in the airway epithelium, lamina propria and ASM in people with asthma (Kruskal–Wallis, p = 0.0018) (where chymase+ cells were present but there were no tryptase+ cells, values of 2 were given). For A‐E) *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 compared with healthy controls (Dunnett's or Dunn's multiple comparison tests as appropriate). For F) ##p < 0.01 compared with airway epithelium (Dunn's multiple comparison test). Red points represent people taking oral corticosteroids
FIGURE 3
FIGURE 3
Remodelling features in asthma subgroups and healthy controls. A) Airway smooth muscle (ASM) mass expressed as a percentage of total biopsy area (Kruskal–Wallis p = 0.0098). B) Mucosal gland mass expressed as a percentage of total biopsy area (Kruskal–Wallis p = 0.0026). C) MUC5AC immunostaining expressed as a percentage of epithelial area (Kruskal–Wallis p < 0.0001). D) Reticular basement membrane (RBM) thickness (ANOVA p = 0.0910). *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 compared with healthy controls (Dunn's multiple comparison test). For A), #p = 0.0506 compared with healthy (Dunn's multiple comparison test). Red points represent people taking oral corticosteroids
FIGURE 4
FIGURE 4
T2‐associated cytokine/chemokine, LTE4 and PGD2 concentrations in induced sputum supernatants in asthma subgroups and healthy controls. *p < 0.05, **p < 0.01, compared with healthy controls (Dunn's multiple comparison test). #p < 0.05 compared with T2‐low (Dunn's multiple comparison test). Red points represent people taking oral corticosteroids. Dotted lines represent the lower limit of detection

References

    1. Stock S, Redaelli M, Luengen M, Wendland G, Civello D, Lauterbach KW. Asthma: prevalence and cost of illness. Eur Respir J. 2005;25(1):47‐53.
    1. Wenzel S. Severe asthma in adults. Am J Respir Crit Care Med. 2005;172(2):149‐160.
    1. Proceedings of the ATS workshop on refractory asthma . Current understanding, recommendations, and unanswered questions. Am J Respir Crit Care Med. 2000;162(6):2341‐2351.
    1. Berry MA, Morgan A, Shaw DE, et al. Pathological features and inhaled corticosteroid response of eosinophilic and non‐eosinophilic asthma. Thorax. 2007;62:1043‐1049.
    1. McGrath KW, Icitovic N, Boushey HA, et al. A large subgroup of mild‐to‐moderate asthma is persistently noneosinophilic. Am J Respir Crit Care Med. 2012;185(6):612‐619.
    1. Heaney LG, Busby J, Hanratty CE, et al. Composite type‐2 biomarker strategy versus a symptom‐risk‐based algorithm to adjust corticosteroid dose in patients with severe asthma: a multicentre, single‐blind, parallel group, randomised controlled trial. Lancet Respir Med. 2020;9:57‐68.
    1. Jackson DJ, Busby J, Pfeffer PE, et al. Characterisation of patients with severe asthma in the UKsevere asthma registry in the biologic era. Thorax. 2021;76(3):220‐227.
    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. Choy DF, Modrek B, Abbas AR, et al. Gene expression patterns of Th2 inflammation and intercellular communication in asthmatic airways. J Immunol. 2010;186(3):1861‐1869. doi:10.4049/jimmunol.1002568
    1. Choy DF, Hart KM, Borthwick LA, et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci Transl Med. 2015;7(301):301ra129.
    1. Ostling J, van Geest M, Schofield JPR, et al. IL‐17‐high asthma with features of a psoriasis immunophenotype. J Allergy Clin Immunol. 2019;144(5):1198‐1213.
    1. Flood‐Page P, Menzies‐Gow A, Phipps S, et al. Anti‐IL‐5 treatment reduces deposition of ECM proteins in the bronchial subepithelial basement membrane of mild atopic asthmatics. J Clin Invest. 2003;112(7):1029‐1036.
    1. Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368(26):2455‐2466.
    1. Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360(10):973‐984.
    1. Hinks TS, Zhou X, Staples KJ, et al. Innate and adaptive T cells in asthmatic patients: relationship to severity and disease mechanisms. J Allergy Clin Immunol. 2015;136(2):323‐333.
    1. Shikotra A, Choy DF, Ohri CM, et al. Increased expression of immunoreactive thymic stromal lymphopoietin in patients with severe asthma. J Allergy Clin Immunol. 2011;129:104‐111.
    1. Siddiqui S, Shikotra A, Richardson M, et al. Airway pathological heterogeneity in asthma: visualization of disease microclusters using topological data analysis. J Allergy Clin Immunol. 2018;142(5):1457‐1468.
    1. Wilson SJ, Ward JA, Sousa AR, et al. Severe asthma exists despite suppressed tissue inflammation: findings of the U‐BIOPRED study. Eur Respir J. 2016;48(5):1307‐1319.
    1. Raundhal M, Morse C, Khare A, et al. High IFN‐gamma and low SLPI mark severe asthma in mice and humans. J Clin Invest. 2015;125(8):3037‐3050.
    1. Kuo CS, Pavlidis S, Loza M, et al. T‐helper cell type 2 (Th2) and non‐Th2 molecular phenotypes of asthma using sputum transcriptomics in U‐BIOPRED. Eur Respir J. 2017;49(2):1602135.
    1. Seys SF, Grabowski M, Adriaensen W, et al. Sputum cytokine mapping reveals an 'IL‐5, IL‐17A, IL‐25‐high' pattern associated with poorly controlled asthma. Clin Exp Allergy. 2013;43(9):1009‐1017.
    1. Austin CD, Gonzalez Edick M, Ferrando RE, et al. A randomized, placebo‐controlled trial evaluating effects of lebrikizumab on airway eosinophilic inflammation and remodelling in uncontrolled asthma (CLAVIER). Clin Exp Allergy. 2020;50(12):1342‐1351.
    1. Heaney LG, Busby J, Bradding P, et al. Remotely monitored therapy and nitric oxide suppression identifies nonadherence in severe asthma. Am J Respir Crit Care Med. 2019;199(4):454‐464.
    1. Boddy CE, Naveed S, Craner M, Murphy AC, Siddiqui S, Bradding P. Clinical outcomes in people with difficult‐to‐control asthma using electronic monitoring to support medication adherence. J Allergy Clin Immunol Pract. 2020;9:1529‐1538.
    1. McNicholl DM, Stevenson M, McGarvey LP, Heaney LG. The utility of fractional exhaled nitric oxide suppression in the identification of nonadherence in difficult asthma. Am J Respir Crit Care Med. 2012;186(11):1102‐1108.
    1. Du Rand IA, Blaikley J, Booton R, et al. British Thoracic Society guideline for diagnostic flexible bronchoscopy in adults: accredited by NICE. Thorax. 2013;68(Suppl 1):i1‐i44.
    1. Sullivan P, Stephens D, Ansari T, Costello J, Jeffery P. Variation in the measurements of basement membrane thickness and inflammatory cell number in bronchial biopsies. Eur Respir J. 1998;12(4):811‐815.
    1. Couillard S, Shrimanker R, Chaudhuri R, et al. FeNO non‐suppression identifies corticosteroid‐resistant Type‐2 signaling in severe asthma. Am J Respir Crit Care Med. 2021;204:731‐734.
    1. Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double‐blind, placebo‐controlled trial. Lancet. 2012;380(9842):651‐659.
    1. Hartl S, Breyer MK, Burghuber OC, et al. Blood eosinophil count in the general population: typical values and potential confounders. Eur Respir J. 2020;55(5):1901874.
    1. Vrugt B, Wilson S, Underwood J, et al. Mucosal inflammation in severe glucocorticoid‐dependent asthma. Eur Respir J. 1999;13(6):1245‐1252.
    1. Wenzel SE, Schwartz LB, Langmack EL, et al. Evidence that severe asthma can be divided pathologically into two inflammatory subtypes with distinct physiologic and clinical characteristics. Am J Respirat Crit Care Med. 1999;160(3):1001‐1008.
    1. Djukanovic R, Wilson JW, Britten KM, et al. Effect of an inhaled corticosteroid on airway inflammation and symptoms in asthma. Am Rev Respir Dis. 1992;145(3):669‐674.
    1. Laitinen LA, Laitinen A, Haahtela T. A comparative study of the effects of an inhaled corticosteroid, budesonide, and a beta 2‐agonist, terbutaline, on airway inflammation in newly diagnosed asthma: a randomized, double‐blind, parallel‐group controlled trial. J Allergy Clin Immunol. 1992;90(1):32‐42.
    1. Kelly EA, Esnault S, Liu LY, et al. Mepolizumab attenuates airway eosinophil numbers, but not their functional phenotype, in asthma. Am J Respir Crit Care Med. 2017;196(11):1385‐1395.
    1. Castro M, Corren J, Pavord ID, et al. Dupilumab efficacy and safety in moderate‐to‐severe uncontrolled asthma. N Engl J Med. 2018;378(26):2486‐2496.
    1. Ortega HG, Liu MC, Pavord ID, et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N Engl J Med. 2014;371(13):1198‐1207.
    1. Ortega HG, Yancey SW, Mayer B, et al. Severe eosinophilic asthma treated with mepolizumab stratified by baseline eosinophil thresholds: a secondary analysis of the DREAM and MENSA studies. Lancet Respir Med. 2016;4(7):549‐556.
    1. Mukherjee M, Aleman Paramo F, Kjarsgaard M, et al. Weight‐adjusted intravenous Reslizumab in severe asthma with inadequate response to fixed‐dose subcutaneous mepolizumab. Am J Respir Crit Care Med. 2018;197(1):38‐46.
    1. Venkayya R, Lam M, Willkom M, Grunig G, Corry DB, Erle DJ. The Th2 lymphocyte products IL‐4 and IL‐13 rapidly induce airway hyperresponsiveness through direct effects on resident airway cells. Am J Respirat Cell Mol Biol. 2002;26(2):202‐208.
    1. Manson ML, Safholm J, James A, et al. IL‐13 and IL‐4, but not IL‐5 nor IL‐17A, induce hyperresponsiveness in isolated human small airways. J Allergy Clin Immunol. 2020;145(3):808‐817. e802.
    1. McDowell PJ, Diver S, Yang F, et al. The inflammatory profile of exacerbations in patients with severe refractory eosinophilic asthma receiving mepolizumab (the MEX study): a prospective observational study. Lancet Respir Med. 2021;9:1174‐1184.
    1. Benayoun L, Druilhe A, Dombret MC, Aubier M, Pretolani M. Airway structural alterations selectively associated with severe asthma. Am J Respir Crit Care Med. 2003;167(10):1360‐1368.
    1. Cutz E, Levison H, Cooper DM. Ultrastructure of airways in children with asthma. Histopathology. 1978;2(6):407‐421.
    1. Huber HL, Koessler KK. The pathology of bronchial asthma. Arch Intern Med. 1922;30:689‐695.
    1. Carroll NG, Mutavdzic S, James AL. Increased mast cells and neutrophils in submucosal mucous glands and mucus plugging in patients with asthma. Thorax. 2002;57(8):677‐682.
    1. Dunican EM, Elicker BM, Gierada DS, et al. Mucus plugs in patients with asthma linked to eosinophilia and airflow obstruction. J Clin Invest. 2018;128(3):997‐1009.
    1. Ordonez CL, Khashayar R, Wong HH, et al. Mild and moderate asthma is associated with airway goblet cell hyperplasia and abnormalities in mucin gene expression. Am J Respir Crit Care Med. 2001;163(2):517‐523.
    1. Kuyper LM, Pare PD, Hogg JC, et al. Characterization of airway plugging in fatal asthma. Am J Med. 2003;115(1):6‐11.
    1. Dunnill MS. The pathology of asthma with special reference to changes in the bronchial mucosa. J Clin Pathol. 1960;13:27‐33.
    1. Thai P, Loukoianov A, Wachi S, Wu R. Regulation of airway mucin gene expression. Annu Rev Physiol. 2008;70:405‐429.
    1. Okumura S, Sagara H, Fukuda T, Saito H, Okayama Y. FcepsilonRI‐mediated amphiregulin production by human mast cells increases mucin gene expression in epithelial cells. J Allergy Clin Immunol. 2005;115(2):272‐279.
    1. Arthur GK, Duffy SM, Roach KM, et al. KCa3.1 K+ channel expression and function in human bronchial epithelial cells. PLoS One. 2015;10(12):e0145259.
    1. Bradding P, Arthur G. Mast cells in asthma‐‐state of the art. Clin Exp Allergy. 2016;46(2):194‐263.
    1. Tiotiu A, Badi Y, Kermani NZ, et al. Association of differential mast cell activation to granulocytic inflammation in severe asthma. Am J Respir Crit Care Med. 2022;205(4):397‐411.
    1. Bradding P, Roberts JA, Britten KM, et al. Interleukin‐4, −5, and −6 and tumor necrosis factor‐alpha in normal and asthmatic airways: evidence for the human mast cell as a source of these cytokines. Am J Respir Cell Mol Biol. 1994;10(5):471‐480.
    1. Wallin A, Sandstrom T, Soderberg M, et al. The effects of regular inhaled formoterol, budesonide, and placebo on mucosal inflammation and clinical indices in mild asthma. Am J Respir Crit Care Med. 1999;159(1):79‐86.
    1. Humbert M, de Blay F, Garcia G, et al. Masitinib, a c‐kit/PDGF receptor tyrosine kinase inhibitor, improves disease control in severe corticosteroid‐dependent asthmatics. Allergy. 2009;64(8):1194‐1201.
    1. Davidescu L, Chanez P, Ursol G, et al. Late Breaking Abstract ‐ Masitinib in severe asthma: Results from a randomized, phase 3 trial. Eur Respir J. 2020;56:4612.
    1. Brightling CE, Bradding P, Symon FA, Holgate ST, Wardlaw AJ, Pavord ID. Mast cell infiltration of airway smooth muscle in asthma. N Engl J Med. 2002;346:1699‐1705.
    1. Begueret H, Berger P, Vernejoux JM, Dubuisson L, Marthan R, Tunon‐De‐Lara JM. Inflammation of bronchial smooth muscle in allergic asthma. Thorax. 2007;62(1):8‐15.
    1. Djukanovic R, Wilson JW, Britten KM, et al. Quantitation of mast cells and eosinophils in the bronchial mucosa of symptomatic atopic asthmatics and healthy control subjects using immunohistochemistry. Am Rev Respir Dis. 1990;142(4):863‐871.
    1. Roberts JA, Bradding P, Britten KM, et al. The long‐acting beta2‐agonist salmeterol xinafoate: effects on airway inflammation in asthma. Eur Respir J. 1999;14(2):275‐282.
    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. Nair P, Surette MG, Virchow JC. Neutrophilic asthma: Misconception or misnomer? Lancet Respir Med. 2021;9:441‐443.
    1. Shikotra A, Choy DF, Siddiqui S, et al. A CEACAM6‐high airway neutrophil phenotype and CEACAM6‐high epithelial cells are features of severe asthma. J Immunol. 2017;198(8):3307‐3317.
    1. Singhania A, Rupani H, Jayasekera N, et al. Altered epithelial gene expression in peripheral airways of severe asthma. PLoS One. 2017;12(1):e0168680.
    1. Kelsen SG, Agache IO, Soong W, et al. Astegolimab (anti‐ST2) efficacy and safety in adults with severe asthma: a randomized clinical trial. J Allergy Clin Immunol. 2021;148(3):790‐798.

Source: PubMed

3
Abonner