184
Views
15
CrossRef citations to date
0
Altmetric
Review

Recent Insights into the Management of Inflammation in Asthma

, , ORCID Icon & ORCID Icon
Pages 4371-4397 | Published online: 02 Sep 2021

References

  • Global Initiative for Asthma. Global strategy for asthma management and prevention; 2021. Available from: www.ginasthma.org. Accessed August 17, 2021.
  • Rackemann FM. A working classification of asthma. Am J Med. 1947;3(5):601–606. doi:10.1016/0002-9343(47)90204-0
  • Brown HM. Treatment of chronic asthma with prednisolone; significance of eosinophils in the sputum. Lancet. 1958;272(7059):1245–1247. doi:10.1016/S0140-6736(58)91385-0
  • 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 Respir Crit Care Med. 1999;160(3):1001–1008. doi:10.1164/ajrccm.160.3.9812110
  • 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. doi:10.1164/rccm.200903-0392OC
  • Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol. 2015;16(1):45–56. doi:10.1038/ni.3049
  • Lambrecht BN, Hammad H, Fahy JV. The cytokines of asthma. Immunity. 2019;50(4):975–991. doi:10.1016/j.immuni.2019.03.018
  • Ingram JL, Kraft M. IL-13 in asthma and allergic disease: asthma phenotypes and targeted therapies. J Allergy Clin Immunol. 2012;130(4):829–842; quiz 843–824. doi:10.1016/j.jaci.2012.06.034
  • 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 Invest. 1999;103(6):779–788. doi:10.1172/JCI5909
  • Pelaia C, Paoletti G, Puggioni F, et al. Interleukin-5 in the pathophysiology of severe asthma. Front Physiol. 2019;10:1514. doi:10.3389/fphys.2019.01514
  • Fitzpatrick AM, Chipps BE, Holguin F, Woodruff PG. T2-”low” asthma: overview and management strategies. J Allergy Clin Immunol Pract. 2020;8(2):452–463. doi:10.1016/j.jaip.2019.11.006
  • Rakowski E, Zhao S, Liu M, et al. Variability of blood eosinophils in patients in a clinic for severe asthma. Clin Exp Allergy. 2019;49(2):163–170. doi:10.1111/cea.13310
  • Jatakanon A, Uasuf C, Maziak W, Lim S, Chung KF, Barnes PJ. Neutrophilic inflammation in severe persistent asthma. Am J Respir Crit Care Med. 1999;160(5):1532–1539. doi:10.1164/ajrccm.160.5.9806170
  • Agache I, Ciobanu C, Agache C, Anghel M. Increased serum IL-17 is an independent risk factor for severe asthma. Respir Med. 2010;104(8):1131–1137. doi:10.1016/j.rmed.2010.02.018
  • Al-Ramli W, Prefontaine D, Chouiali F, et al. T(H)17-associated cytokines (IL-17A and IL-17F) in severe asthma. J Allergy Clin Immunol. 2009;123(5):1185–1187. doi:10.1016/j.jaci.2009.02.024
  • Tliba O, Panettieri RA. Paucigranulocytic asthma: uncoupling of airway obstruction from inflammation. J Allergy Clin Immunol. 2019;143(4):1287–1294. doi:10.1016/j.jaci.2018.06.008
  • Peters MC, McGrath KW, Hawkins GA, et al. Plasma interleukin-6 concentrations, metabolic dysfunction, and asthma severity: a cross-sectional analysis of two cohorts. Lancet Respir Med. 2016;4(7):574–584. doi:10.1016/S2213-2600(16)30048-0
  • (GINA) GIFA. Difficult-to-treat and severe asthma in adolescent and adult patients: diagnosis and management. 2019.
  • (GINA) GIFA. Global strategy for asthma management and prevention. 2020.
  • Jackson DJ, Busby J, Pfeffer PE, et al. Characterisation of patients with severe asthma in the UK severe asthma registry in the biologic era. Thorax. 2021;76(3):220–227. doi:10.1136/thoraxjnl-2020-215168
  • Heaney LG, Perez de Llano L, Al-Ahmad M, et al. Eosinophilic and noneosinophilic asthma: an expert consensus framework to characterize phenotypes in a global real-life severe asthma cohort. Chest. 2021:In Press. doi:10.1016/j.chest.2021.04.013
  • Azim A, Newell C, Barber C, et al. Clinical evaluation of type 2 disease status in a real-world population of difficult to manage asthma using historic electronic healthcare records of blood eosinophil counts. Clin Exp Allergy. 2021;51(6):811–820. doi:10.1111/cea.13841
  • Azim A, Freeman A, Lavenu A, et al. New perspectives on difficult asthma; sex and age of asthma-onset based phenotypes. J Allergy Clin Immunol Pract. 2020;8(10):3396–3406 e3394. doi:10.1016/j.jaip.2020.05.053
  • Lefaudeux D, De Meulder B, Loza MJ, et al. U-BIOPRED clinical adult asthma clusters linked to a subset of sputum omics. J Allergy Clin Immunol. 2017;139(6):1797–1807. doi:10.1016/j.jaci.2016.08.048
  • 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. doi:10.1164/rccm.200906-0896OC
  • Fong WCG, Azim A, Knight D, et al. Real-world Omalizumab and Mepolizumab treated difficult asthma phenotypes and their clinical outcomes. Clin Exp Allergy. 2021;51:1019–1032. doi:10.1111/cea.13882
  • Molina C, Brun J, Coulet M, Betail G, Delage J. Immunopathology of the bronchial mucosa in ‘late onset’ asthma. Clin Allergy. 1977;7(2):137–145. doi:10.1111/j.1365-2222.1977.tb01434.x
  • 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. doi:10.1164/ajrccm/142.4.863
  • Jarjour NN, Peters SP, Djukanovic R, Calhoun WJ. Investigative use of bronchoscopy in asthma. Am J Respir Crit Care Med. 1998;157(3):692–697. doi:10.1164/ajrccm.157.3.9705020
  • Simpson JL, Scott R, Boyle MJ, Gibson PG. Inflammatory subtypes in asthma: assessment and identification using induced sputum. Respirology. 2006;11(1):54–61. doi:10.1111/j.1440-1843.2006.00784.x
  • Louis R, Sele J, Henket M, et al. Sputum eosinophil count in a large population of patients with mild to moderate steroid-naive asthma: distribution and relationship with methacholine bronchial hyperresponsiveness. Allergy. 2002;57(10):907–912. doi:10.1034/j.1398-9995.2002.23608.x
  • Green RH, Brightling CE, McKenna S, et al. Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet. 2002;360(9347):1715–1721. doi:10.1016/S0140-6736(02)11679-5
  • Fleming L, Wilson N, Regamey N, Bush A. Use of sputum eosinophil counts to guide management in children with severe asthma. Thorax. 2012;67(3):193–198. doi:10.1136/thx.2010.156836
  • Jones TB, Elliott T, Rupani S, et al. Characteristics of eosinophilic severe asthmatics in the Wessex Severe Asthma Cohort (WSAC). Eur Respir J. 2017;50:PA4042.
  • Ricciardolo FL. Revisiting the role of exhaled nitric oxide in asthma. Curr Opin Pulm Med. 2014;20(1):53–59. doi:10.1097/MCP.0000000000000006
  • Chibana K, Trudeau JB, Mustovich AT, et al. IL-13 induced increases in nitrite levels are primarily driven by increases in inducible nitric oxide synthase as compared with effects on arginases in human primary bronchial epithelial cells. Clin Exp Allergy. 2008;38(6):936–946. doi:10.1111/j.1365-2222.2008.02969.x
  • Ichinose M, Sugiura H, Yamagata S, Koarai A, Shirato K. Increase in reactive nitrogen species production in chronic obstructive pulmonary disease airways. Am J Respir Crit Care Med. 2000;162(2):701–706. doi:10.1164/ajrccm.162.2.9908132
  • Kupczyk M, Ten Brinke A, Sterk PJ, et al. Frequent exacerbators–a distinct phenotype of severe asthma. Clin Exp Allergy. 2014;44(2):212–221. doi:10.1111/cea.12179
  • Coumou H, Westerhof GA, de Nijs SB, Zwinderman AH, Bel EH. Predictors of accelerated decline in lung function in adult-onset asthma. Eur Respir J. 2018;51(2):1701785. doi:10.1183/13993003.01785-2017
  • Matsunaga K, Hirano T, Oka A, Ito K, Edakuni N. Persistently high exhaled nitric oxide and loss of lung function in controlled asthma. Allergol Int. 2016;65(3):266–271. doi:10.1016/j.alit.2015.12.006
  • Vijverberg SJ, Hilvering B, Raaijmakers JA, Lammers JW, Maitland-van der Zee AH, Koenderman L. Clinical utility of asthma biomarkers: from bench to bedside. Biologics. 2013;7:199–210.
  • Dweik RA, Boggs PB, Erzurum SC, et al. An official ATS clinical practice guideline: interpretation of exhaled nitric oxide levels (FENO) for clinical applications. Am J Respir Crit Care Med. 2011;184(5):602–615. doi:10.1164/rccm.9120-11ST
  • Price DB, Buhl R, Chan A, et al. Fractional exhaled nitric oxide as a predictor of response to inhaled corticosteroids in patients with non-specific respiratory symptoms and insignificant bronchodilator reversibility: a randomised controlled trial. Lancet Respir Med. 2018;6(1):29–39. doi:10.1016/S2213-2600(17)30424-1
  • Cloutier MM, Baptist AP, Blake KV, et al. 2020 focused updates to the asthma management guidelines: a report from the National Asthma Education and Prevention Program Coordinating Committee Expert Panel Working Group. J Allergy Clin Immunol. 2020;146(6):1217–1270. doi:10.1016/j.jaci.2020.10.003
  • 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. doi:10.1164/rccm.201204-0587OC
  • Petsky HL, Cates CJ, Kew KM, Chang AB. Tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or sputum eosinophils): a systematic review and meta-analysis. Thorax. 2018;73(12):1110–1119. doi:10.1136/thoraxjnl-2018-211540
  • Korevaar DA, Westerhof GA, Wang J, et al. Diagnostic accuracy of minimally invasive markers for detection of airway eosinophilia in asthma: a systematic review and meta-analysis. Lancet Respir Med. 2015;3(4):290–300. doi:10.1016/S2213-2600(15)00050-8
  • Castro M, Corren J, Pavord ID, et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N Engl JMed. 2018;378:2486–2496. doi:10.1056/NEJMoa1804092
  • 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. doi:10.1016/S0140-6736(12)60988-X
  • Wagener AH, de Nijs SB, Lutter R, et al. External validation of blood eosinophils, FE(NO) and serum periostin as surrogates for sputum eosinophils in asthma. Thorax. 2015;70(2):115–120. doi:10.1136/thoraxjnl-2014-205634
  • Hancox RJ, Pavord ID, Sears MR. Associations between blood eosinophils and decline in lung function among adults with and without asthma. Eur Respir J. 2018;51(4):1702536. doi:10.1183/13993003.02536-2017
  • Tan WC, Bourbeau J, Nadeau G, et al. High eosinophil counts predict decline in FEV1: results from the CanCOLD study. Eur Respir J. 2020;57:2000838.
  • Price D, Wilson AM, Chisholm A, et al. Predicting frequent asthma exacerbations using blood eosinophil count and other patient data routinely available in clinical practice. JAA. 2016;9:1–12.
  • Shrimanker R, Keene O, Hynes G, Wenzel S, Yancey S, Pavord ID. Prognostic and predictive value of blood eosinophil count, fractional exhaled nitric oxide, and their combination in severe asthma: a post hoc analysis. Am J Respir Crit Care Med. 2019;200(10):1308–1312. doi:10.1164/rccm.201903-0599LE
  • Price DB, Rigazio A, Campbell JD, et al. Blood eosinophil count and prospective annual asthma disease burden: a UK cohort study. Lancet Respir Med. 2015;3(11):849–858. doi:10.1016/S2213-2600(15)00367-7
  • Bleecker ER, Wechsler ME, FitzGerald JM, et al. Baseline patient factors impact on the clinical efficacy of benralizumab for severe asthma. Eur Respir J. 2018;52(4):1800936. doi:10.1183/13993003.00936-2018
  • Licari A, Manti S, Castagnoli R, Leonardi S, Marseglia GL. Measuring inflammation in paediatric severe asthma: biomarkers in clinical practice. Breathe. 2020;16(1):190301. doi:10.1183/20734735.0301-2019
  • Pavord ID, Afzalnia S, Menzies-Gow A, Heaney LG. The current and future role of biomarkers in type 2 cytokine-mediated asthma management. Clin Exp Allergy. 2017;47(2):148–160. doi:10.1111/cea.12881
  • Takayama G, Arima K, Kanaji T, et al. Periostin: a novel component of subepithelial fibrosis of bronchial asthma downstream of IL-4 and IL-13 signals. J Allergy Clin Immunol. 2006;118(1):98–104. doi:10.1016/j.jaci.2006.02.046
  • Woodruff PG, Boushey HA, Dolganov GM, et al. Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci USA. 2007;104(40):15858–15863. doi:10.1073/pnas.0707413104
  • 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. doi:10.1016/j.jaci.2012.06.025
  • Couillard S, Laugerud A, Jabeen M, et al. A proof-of-concept scale to predict asthma attacks: the OxfoRd Asthma attaCk risk ScaLE (ORACLE). Am J Respir Crit Care Med. 2021;203:A1436.
  • Parikh NI, Vasan RS. Assessing the clinical utility of biomarkers in medicine. Biomark Med. 2007;1(3):419–436. doi:10.2217/17520363.1.3.419
  • Gebert LFR, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. 2019;20(1):21–37.
  • Specjalski K, Niedoszytko M. MicroRNAs: future biomarkers and targets of therapy in asthma? Curr Opin Pulm Med. 2020;26(3):285–292. doi:10.1097/MCP.0000000000000673
  • Huang Y, Zhang S, Fang X, et al. Plasma miR-199a-5p is increased in neutrophilic phenotype asthma patients and negatively correlated with pulmonary function. PLoS One. 2018;13(3):e0193502. doi:10.1371/journal.pone.0193502
  • Tian M, Zhou Y, Jia H, Zhu X, Cui Y. The clinical significance of changes in the expression levels of MicroRNA-1 and inflammatory factors in the peripheral blood of children with acute-stage asthma. Biomed Res Int. 2018;2018:7632487. doi:10.1155/2018/7632487
  • Maes T, Cobos FA, Schleich F, et al. Asthma inflammatory phenotypes show differential microRNA expression in sputum. J Allergy Clin Immunol. 2016;137(5):1433–1446. doi:10.1016/j.jaci.2016.02.018
  • Zhao M, Juanjuan L, Weijia F, et al. Expression levels of microRNA-125b in serum exosomes of patients with asthma of different severity and its diagnostic significance. Curr Drug Metab. 2019;20(10):781–784. doi:10.2174/1389200220666191021100001
  • Rodrigo-Munoz JM, Canas JA, Sastre B, et al. Asthma diagnosis using integrated analysis of eosinophil microRNAs. Allergy. 2019;74(3):507–517. doi:10.1111/all.13570
  • de Vries R, Dagelet YWF, Spoor P, et al. Clinical and inflammatory phenotyping by breathomics in chronic airway diseases irrespective of the diagnostic label. Eur Respir J. 2018;51(1):1701817. doi:10.1183/13993003.01817-2017
  • Schleich FN, Zanella D, Stefanuto PH, et al. Exhaled volatile organic compounds are able to discriminate between neutrophilic and eosinophilic asthma. Am J Respir Crit Care Med. 2019;200(4):444–453. doi:10.1164/rccm.201811-2210OC
  • Brinkman P, van de Pol MA, Gerritsen MG, et al. Exhaled breath profiles in the monitoring of loss of control and clinical recovery in asthma. Clin Exp Allergy. 2017;47(9):1159–1169. doi:10.1111/cea.12965
  • Peel AM, Wilkinson M, Sinha A, Loke YK, Fowler SJ, Wilson AM. Volatile organic compounds associated with diagnosis and disease characteristics in asthma - a systematic review. Respir Med. 2020;169:105984. doi:10.1016/j.rmed.2020.105984
  • Holz O, Waschki B, Watz H, et al. Breath volatile organic compounds and inflammatory markers in adult asthma patients: negative results from the ALLIANCE cohort. Eur Respir J. 2021;57(2):2002127. doi:10.1183/13993003.02127-2020
  • Kolmert J, Gomez C, Balgoma D, et al. Urinary leukotriene E4 and prostaglandin D2 metabolites increase in adult and childhood severe asthma characterized by type 2 inflammation. A clinical observational study. Am J Respir Crit Care Med. 2021;203(1):37–53. doi:10.1164/rccm.201909-1869OC
  • Hagan JB, Laidlaw TM, Divekar R, et al. Urinary leukotriene E4 to determine aspirin intolerance in asthma: a systematic review and meta-analysis. J Allergy Clin Immunol Pract. 2017;5(4):990–997 e991. doi:10.1016/j.jaip.2016.11.004
  • 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. doi:10.1016/j.jaci.2019.03.027
  • Cowan DC, Taylor DR, Peterson LE, et al. Biomarker-based asthma phenotypes of corticosteroid response. J Allergy Clin Immunol. 2015;135(4):877–883 e871. doi:10.1016/j.jaci.2014.10.026
  • Denton E, Price DB, Tran TN, et al. Cluster analysis of inflammatory biomarker expression in the international severe asthma registry. J Allergy Clin Immunol Pract. 2021;9:2680–2688.e7. doi:10.1016/j.jaip.2021.02.059
  • Fitzpatrick AM, Teague WG, Meyers DA, et al. Heterogeneity of severe asthma in childhood: confirmation by cluster analysis of children in the National Institutes of Health/National Heart, Lung, and Blood Institute Severe Asthma Research Program. J Allergy Clin Immunol. 2011;127(2):382–389. doi:10.1016/j.jaci.2010.11.015
  • Kurukulaaratchy RJ, Zhang H, Raza A, et al. The diversity of young adult wheeze: a cluster analysis in a longitudinal birth cohort. Clin Exp Allergy. 2014;44(5):724–735. doi:10.1111/cea.12306
  • Pavord ID, Beasley R, Agusti A, et al. After asthma: redefining airways diseases. Lancet. 2018;391(10118):350–400.
  • Agusti A, Bel E, Thomas M, et al. Treatable traits: toward precision medicine of chronic airway diseases. Eur Respir J. 2016;47(2):410–419. doi:10.1183/13993003.01359-2015
  • Freitas PD, Xavier RF, McDonald VM, et al. Identification of asthma phenotypes based on extrapulmonary treatable traits. Eur Respir J. 2021;57(1):2000240. doi:10.1183/13993003.00240-2020
  • McDonald VM, Hiles SA, Godbout K, et al. Treatable traits can be identified in a severe asthma registry and predict future exacerbations. Respirology. 2019;24(1):37–47. doi:10.1111/resp.13389
  • Simpson AJ, Hekking PP, Shaw DE, et al. Treatable traits in the European U-BIOPRED adult asthma cohorts. Allergy. 2019;74(2):406–411. doi:10.1111/all.13629
  • Tay TR, Radhakrishna N, Hore-Lacy F, et al. Comorbidities in difficult asthma are independent risk factors for frequent exacerbations, poor control and diminished quality of life. Respirology. 2016;21(8):1384–1390. doi:10.1111/resp.12838
  • McDonald VM, Clark VL, Cordova-Rivera L, Wark PAB, Baines KJ, Gibson PG. Targeting treatable traits in severe asthma: a randomised controlled trial. Eur Respir J. 2020;55(3):1901509. doi:10.1183/13993003.01509-2019
  • 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 Respir Crit Care Med. 2003;167(2):199–204. doi:10.1164/rccm.200208-789OC
  • Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009;360(10):973–984. doi:10.1056/NEJMoa0808991
  • Nair P, Pizzichini MM, Kjarsgaard M, et al. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N Engl J Med. 2009;360(10):985–993. doi:10.1056/NEJMoa0805435
  • Green RH, Brightling CE, Woltmann G, Parker D, Wardlaw AJ, Pavord ID. Analysis of induced sputum in adults with asthma: identification of subgroup with isolated sputum neutrophilia and poor response to inhaled corticosteroids. Thorax. 2002;57(10):875–879. doi:10.1136/thorax.57.10.875
  • Moore WC, Hastie AT, Li X, et al. Sputum neutrophil counts are associated with more severe asthma phenotypes using cluster analysis. J Allergy Clin Immunol. 2014;133(6):1557–1563 e1555. doi:10.1016/j.jaci.2013.10.011
  • Shaw DE, Berry MA, Hargadon B, et al. Association between neutrophilic airway inflammation and airflow limitation in adults with asthma. Chest. 2007;132(6):1871–1875. doi:10.1378/chest.07-1047
  • Boulet LP, Lemiere C, Archambault F, Carrier G, Descary MC, Deschesnes F. Smoking and asthma: clinical and radiologic features, lung function, and airway inflammation. Chest. 2006;129(3):661–668. doi:10.1378/chest.129.3.661
  • Green BJ, Wiriyachaiporn S, Grainge C, et al. Potentially pathogenic airway bacteria and neutrophilic inflammation in treatment resistant severe asthma. PLoS One. 2014;9(6):e100645. doi:10.1371/journal.pone.0100645
  • McCreanor J, Cullinan P, Nieuwenhuijsen MJ, et al. Respiratory effects of exposure to diesel traffic in persons with asthma. N Engl J Med. 2007;357(23):2348–2358. doi:10.1056/NEJMoa071535
  • Hastie AT, Mauger DT, Denlinger LC, et al. Mixed sputum granulocyte longitudinal impact on lung function in the severe asthma research program. Am J Respir Crit Care Med. 2021;203(7):882–892. doi:10.1164/rccm.202009-3713OC
  • Hastie AT, Moore WC, Meyers DA, et al. Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes. J Allergy Clin Immunol. 2010;125(5):1028–1036 e1013. doi:10.1016/j.jaci.2010.02.008
  • Ntontsi P, Loukides S, Bakakos P, et al. Clinical, functional and inflammatory characteristics in patients with paucigranulocytic stable asthma: comparison with different sputum phenotypes. Allergy. 2017;72(11):1761–1767. doi:10.1111/all.13184
  • Svenningsen S, Nair P. Asthma endotypes and an overview of targeted therapy for asthma. Front Med (Lausanne). 2017;4:158. doi:10.3389/fmed.2017.00158
  • Shin B, Kwon HS, Park SY, Kim TB, Moon HB, Cho YS. The transition of sputum inflammatory cell profiles is variable in stable asthma patients. Asia Pac Allergy. 2017;7(1):19–28. doi:10.5415/apallergy.2017.7.1.19
  • Denning DW, O’Driscoll BR, Hogaboam CM, Bowyer P, Niven RM. The link between fungi and severe asthma: a summary of the evidence. Eur Respir J. 2006;27(3):615–626. doi:10.1183/09031936.06.00074705
  • Rick EM, Woolnough K, Pashley CH, Wardlaw AJ. Allergic fungal airway disease. J Investig Allergol Clin Immunol. 2016;26(6):344–354. doi:10.18176/jiaci.0122
  • Fairs A, Agbetile J, Hargadon B, et al. IgE sensitization to Aspergillus fumigatus is associated with reduced lung function in asthma. Am J Respir Crit Care Med. 2010;182(11):1362–1368. doi:10.1164/rccm.201001-0087OC
  • Goh KJ, Yii ACA, Lapperre TS, et al. Sensitization to Aspergillus species is associated with frequent exacerbations in severe asthma. J Asthma Allergy. 2017;10:131–140. doi:10.2147/JAA.S130459
  • Medrek SK, Kao CC, Yang DH, Hanania NA, Parulekar AD. Fungal sensitization is associated with increased risk of life-threatening asthma. J Allergy Clin Immunol Pract. 2017;5(4):1025–1031 e1022. doi:10.1016/j.jaip.2016.11.015
  • Menzies D, Holmes L, McCumesky G, Prys-Picard C, Niven R. Aspergillus sensitization is associated with airflow limitation and bronchiectasis in severe asthma. Allergy. 2011;66(5):679–685. doi:10.1111/j.1398-9995.2010.02542.x
  • O’Driscoll BR, Hopkinson LC, Denning DW. Mold sensitization is common amongst patients with severe asthma requiring multiple hospital admissions. BMC Pulm Med. 2005;5:4. doi:10.1186/1471-2466-5-4
  • Targonski PV, Persky VW, Ramekrishnan V. Effect of environmental molds on risk of death from asthma during the pollen season. J Allergy Clin Immunol. 1995;95(5):955–961. doi:10.1016/S0091-6749(95)70095-1
  • Woolnough KF, Richardson M, Newby C, et al. The relationship between biomarkers of fungal allergy and lung damage in asthma. Clin Exp Allergy. 2017;47(1):48–56. doi:10.1111/cea.12848
  • Wark PA, Saltos N, Simpson J, Slater S, Hensley MJ, Gibson PG. Induced sputum eosinophils and neutrophils and bronchiectasis severity in allergic bronchopulmonary aspergillosis. Eur Respir J. 2000;16(6):1095–1101. doi:10.1034/j.1399-3003.2000.16f13.x
  • Denning DW, Pashley C, Hartl D, et al. Fungal allergy in asthma-state of the art and research needs. Clin Transl Allergy. 2014;4:14. doi:10.1186/2045-7022-4-14
  • Li JX, Fan LC, Li MH, Cao WJ, Xu JF. Beneficial effects of Omalizumab therapy in allergic bronchopulmonary aspergillosis: a synthesis review of published literature. Respir Med. 2017;122:33–42. doi:10.1016/j.rmed.2016.11.019
  • Wark PA, Gibson PG, Wilson AJ. Azoles for allergic bronchopulmonary aspergillosis associated with asthma. Cochrane Database Syst Rev. 2004;2004(3):CD001108. doi:10.1002/14651858.CD001108.pub2
  • Szczeklik A, Nizankowska E, Duplaga M. Natural history of aspirin-induced asthma. AIANE investigators. European network on aspirin-induced asthma. Eur Respir J. 2000;16(3):432–436. doi:10.1034/j.1399-3003.2000.016003432.x
  • Christie PE, Tagari P, Ford-Hutchinson AW, et al. Urinary leukotriene E4 concentrations increase after aspirin challenge in aspirin-sensitive asthmatic subjects. Am Rev Respir Dis. 1991;143(5 Pt 1):1025–1029. doi:10.1164/ajrccm/143.5_Pt_1.1025
  • White AA, Stevenson DD, Longo DL. Aspirin-exacerbated respiratory disease. N Engl J Med. 2018;379(11):1060–1070. doi:10.1056/NEJMra1712125
  • Adelman J, McLean C, Shaigany K, Krouse JH. The role of surgery in management of Samter’s triad: a systematic review. Otolaryngol Head Neck Surg. 2016;155(2):220–237. doi:10.1177/0194599816640723
  • Sommer DD, Rotenberg BW, Sowerby LJ, et al. A novel treatment adjunct for aspirin exacerbated respiratory disease: the low-salicylate diet: a multicenter randomized control crossover trial. Int Forum Allergy Rhinol. 2016;6(4):385–391. doi:10.1002/alr.21678
  • Waldram J, Walters K, Simon R, Woessner K, Waalen J, White A. Safety and outcomes of aspirin desensitization for aspirin-exacerbated respiratory disease: a single-center study. J Allergy Clin Immunol. 2018;141(1):250–256. doi:10.1016/j.jaci.2017.05.006
  • Hayashi H, Mitsui C, Nakatani E, et al. Omalizumab reduces cysteinyl leukotriene and 9alpha, 11beta-prostaglandin F2 overproduction in aspirin-exacerbated respiratory disease. J Allergy Clin Immunol. 2016;137(5):1585–1587 e1584. doi:10.1016/j.jaci.2015.09.034
  • Bousquet J, Vignola AM, Demoly P. Links between rhinitis and asthma. Allergy. 2003;58(8):691–706. doi:10.1034/j.1398-9995.2003.00105.x
  • de Groot EP, Nijkamp A, Duiverman EJ, Brand PL. Allergic rhinitis is associated with poor asthma control in children with asthma. Thorax. 2012;67(7):582–587. doi:10.1136/thoraxjnl-2011-201168
  • Kurukulaaratchy RJ, Zhang H, Patil V, et al. Identifying the heterogeneity of young adult rhinitis through cluster analysis in the Isle of Wight birth cohort. J Allergy Clin Immunol. 2015;135(1):143–150. doi:10.1016/j.jaci.2014.06.017
  • Scichilone N, Arrigo R, Paterno A, et al. The effect of intranasal corticosteroids on asthma control and quality of life in allergic rhinitis with mild asthma. J Asthma. 2011;48(1):41–47. doi:10.3109/02770903.2010.528821
  • Scadding GK, Kariyawasam HH, Scadding G, et al. BSACI guideline for the diagnosis and management of allergic and non-allergic rhinitis (Revised Edition 2017; First edition 2007). Clin Exp Allergy. 2017;47(7):856–889.
  • Bilodeau L, Boulay ME, Prince P, Boisvert P, Boulet LP. Comparative clinical and airway inflammatory features of asthmatics with or without polyps. Rhinology. 2010;48(4):420–425. doi:10.4193/Rhino09.095
  • Hakansson K, Bachert C, Konge L, et al. Airway inflammation in chronic rhinosinusitis with nasal polyps and asthma: the united airways concept further supported. PLoS One. 2015;10(7):e0127228. doi:10.1371/journal.pone.0127228
  • Jarvis D, Newson R, Lotvall J, et al. Asthma in adults and its association with chronic rhinosinusitis: the GA2LEN survey in Europe. Allergy. 2012;67(1):91–98. doi:10.1111/j.1398-9995.2011.02709.x
  • Bachert C, Zhang N, Patou J, van Zele T, Gevaert P. Role of staphylococcal superantigens in upper airway disease. Curr Opin Allergy Clin Immunol. 2008;8(1):34–38. doi:10.1097/ACI.0b013e3282f4178f
  • Fokkens WJ, Lund VJ, Hopkins C, et al. Executive summary of EPOS 2020 including integrated care pathways. Rhinology. 2020;58(2):82–111. doi:10.4193/Rhin20.601
  • Al Badaai Y, Valdes CJ, Samaha M. Outcomes and cost benefits of functional endoscopic sinus surgery in severely asthmatic patients with chronic rhinosinusitis. J Laryngol Otol. 2014;128(6):512–517. doi:10.1017/S0022215114001133
  • Chen FH, Zuo KJ, Guo YB, et al. Long-term results of endoscopic sinus surgery-oriented treatment for chronic rhinosinusitis with asthma. Laryngoscope. 2014;124(1):24–28. doi:10.1002/lary.24196
  • Ragab S, Scadding GK, Lund VJ, Saleh H. Treatment of chronic rhinosinusitis and its effects on asthma. Eur Respir J. 2006;28(1):68–74. doi:10.1183/09031936.06.00043305
  • Holguin F, Bleecker ER, Busse WW, et al. Obesity and asthma: an association modified by age of asthma onset. J Allergy Clin Immunol. 2011;127(6):1486–1493 e1482. doi:10.1016/j.jaci.2011.03.036
  • Dixon AE, Peters U. The effect of obesity on lung function. Expert Rev Respir Med. 2018;12(9):755–767. doi:10.1080/17476348.2018.1506331
  • Brumpton BM, Camargo CA, Romundstad PR, Langhammer A, Chen Y, Mai XM. Metabolic syndrome and incidence of asthma in adults: the HUNT study. Eur Respir J. 2013;42(6):1495–1502. doi:10.1183/09031936.00046013
  • Forno E, Han YY, Muzumdar RH, Celedon JC. Insulin resistance, metabolic syndrome, and lung function in US adolescents with and without asthma. J Allergy Clin Immunol. 2015;136(2):304–311 e308. doi:10.1016/j.jaci.2015.01.010
  • Kattan M, Kumar R, Bloomberg GR, et al. Asthma control, adiposity, and adipokines among inner-city adolescents. J Allergy Clin Immunol. 2010;125(3):584–592. doi:10.1016/j.jaci.2010.01.053
  • Scott HA, Gibson PG, Garg ML, Wood LG. Airway inflammation is augmented by obesity and fatty acids in asthma. Eur Respir J. 2011;38(3):594–602. doi:10.1183/09031936.00139810
  • Hasegawa K, Tsugawa Y, Chang Y, Camargo CA. Risk of an asthma exacerbation after bariatric surgery in adults. J Allergy Clin Immunol. 2015;136(2):288–294 e288. doi:10.1016/j.jaci.2014.12.1931
  • Nyenhuis SM, Dixon AE, Ma J. Impact of lifestyle interventions targeting healthy diet, physical activity, and weight loss on asthma in adults: what is the evidence? J Allergy Clin Immunol Pract. 2018;6(3):751–763. doi:10.1016/j.jaip.2017.10.026
  • Okoniewski W, Lu KD, Forno E. Weight loss for children and adults with obesity and asthma. A systematic review of randomized controlled trials. Ann Am Thorac Soc. 2019;16(5):613–625. doi:10.1513/AnnalsATS.201810-651SR
  • van Huisstede A, Rudolphus A, Castro Cabezas M, et al. Effect of bariatric surgery on asthma control, lung function and bronchial and systemic inflammation in morbidly obese subjects with asthma. Thorax. 2015;70(7):659–667. doi:10.1136/thoraxjnl-2014-206712
  • Chalmers GW, Macleod KJ, Little SA, Thomson LJ, McSharry CP, Thomson NC. Influence of cigarette smoking on inhaled corticosteroid treatment in mild asthma. Thorax. 2002;57(3):226–230. doi:10.1136/thorax.57.3.226
  • Lazarus SC, Chinchilli VM, Rollings NJ, et al. Smoking affects response to inhaled corticosteroids or leukotriene receptor antagonists in asthma. Am J Respir Crit Care Med. 2007;175(8):783–790. doi:10.1164/rccm.200511-1746OC
  • Chaudhuri R, Livingston E, McMahon AD, et al. Effects of smoking cessation on lung function and airway inflammation in smokers with asthma. Am J Respir Crit Care Med. 2006;174(2):127–133. doi:10.1164/rccm.200510-1589OC
  • Perret JL, Bonevski B, McDonald CF, Abramson MJ. Smoking cessation strategies for patients with asthma: improving patient outcomes. J Asthma Allergy. 2016;9:117–128. doi:10.2147/JAA.S85615
  • Gamble J, Stevenson M, McClean E, Heaney LG. The prevalence of nonadherence in difficult asthma. Am J Respir Crit Care Med. 2009;180(9):817–822. doi:10.1164/rccm.200902-0166OC
  • Murphy AC, Proeschal A, Brightling CE, et al. The relationship between clinical outcomes and medication adherence in difficult-to-control asthma. Thorax. 2012;67(8):751–753. doi:10.1136/thoraxjnl-2011-201096
  • Alahmadi F, Peel A, Keevil B, Niven R, Fowler SJ. Assessment of adherence to corticosteroids in asthma by drug monitoring or fractional exhaled nitric oxide: a literature review. Clin Exp Allergy. 2021;51(1):49–62. doi:10.1111/cea.13787
  • Averell CM, Stanford RH, Laliberte F, Wu JW, Germain G, Duh MS. Medication adherence in patients with asthma using once-daily versus twice-daily ICS/LABAs. J Asthma. 2021;58(1):102–111. doi:10.1080/02770903.2019.1663429
  • 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. doi:10.1164/rccm.201806-1182OC
  • Moore A, Preece A, Sharma R, et al. A randomised controlled trial of the effect of a connected inhaler system on medication adherence in uncontrolled asthmatic patients. Eur Respir J. 2021;57(6):2003103. doi:10.1183/13993003.03103-2020
  • Mosnaim G, Safioti G, Brown R, et al. Digital health technology in asthma: a comprehensive scoping review. J Allergy Clin Immunol Pract. 2021;9:2377–2398. doi:10.1016/j.jaip.2021.02.028
  • Wells KE, Peterson EL, Ahmedani BK, Williams LK. Real-world effects of once vs greater daily inhaled corticosteroid dosing on medication adherence. Ann Allergy Asthma Immunol. 2013;111(3):216–220. doi:10.1016/j.anai.2013.06.008
  • Papi A, Blasi F, Canonica GW, Morandi L, Richeldi L, Rossi A. Treatment strategies for asthma: reshaping the concept of asthma management. Allergy Asthma Clin Immunol. 2020;16:75. doi:10.1186/s13223-020-00472-8
  • Dusser D, Montani D, Chanez P, et al. Mild asthma: an expert review on epidemiology, clinical characteristics and treatment recommendations. Allergy. 2007;62(6):591–604. doi:10.1111/j.1398-9995.2007.01394.x
  • Miller MK, Lee JH, Miller DP, Wenzel SE, Group TS. Recent asthma exacerbations: a key predictor of future exacerbations. Respir Med. 2007;101(3):481–489. doi:10.1016/j.rmed.2006.07.005
  • Suissa S, Ernst P, Benayoun S, Baltzan M, Cai B. Low-dose inhaled corticosteroids and the prevention of death from asthma. N Engl J Med. 2000;343(5):332–336. doi:10.1056/NEJM200008033430504
  • Suissa S, Ernst P, Kezouh A. Regular use of inhaled corticosteroids and the long term prevention of hospitalisation for asthma. Thorax. 2002;57(10):880–884. doi:10.1136/thorax.57.10.880
  • Haahtela T, Tuomisto LE, Pietinalho A, et al. A 10 year asthma programme in Finland: major change for the better. Thorax. 2006;61(8):663. doi:10.1136/thx.2005.055699
  • Barnes CB, Ulrik CS. Asthma and adherence to inhaled corticosteroids: current status and future perspectives. Respir Care. 2015;60(3):455–468. doi:10.4187/respcare.03200
  • Bateman ED, Reddel HK, FitzGerald JM. As-needed budesonide-formoterol in mild asthma. N Engl J Med. 2018;379(9):898.
  • Beasley R, Weatherall M, Shirtcliffe P, Hancox R, Reddel HK. Combination corticosteroid/beta-agonist inhaler as reliever therapy: a solution for intermittent and mild asthma? J Allergy Clin Immunol. 2014;133(1):39–41. doi:10.1016/j.jaci.2013.10.053
  • Price D, Fletcher M, van der Molen T. Asthma control and management in 8000 European patients: the REcognise Asthma and LInk to Symptoms and Experience (REALISE) survey. NPJ Prim Care Respir Med. 2014;24:14009. doi:10.1038/npjpcrm.2014.9
  • O’Byrne PM, FitzGerald JM, Bateman ED, et al. Inhaled combined budesonide-formoterol as needed in mild asthma. N Engl J Med. 2018;378(20):1865–1876. doi:10.1056/NEJMoa1715274
  • Beasley R, Holliday M, Reddel HK, et al. Controlled trial of budesonide–formoterol as needed for mild asthma. N Engl JMed. 2019;380(21):2020–2030. doi:10.1056/NEJMoa1901963
  • Sobieraj DM, Weeda ER, Nguyen E, et al. Association of inhaled corticosteroids and long-acting β-agonists as controller and quick relief therapy with exacerbations and symptom control in persistent asthma: a systematic review and meta-analysis. JAMA. 2018;319(14):1485–1496. doi:10.1001/jama.2018.2769
  • Tattersfield AE, Postma DS, Barnes PJ, et al. Exacerbations of asthma: a descriptive study of 425 severe exacerbations. The FACET International Study Group. Am J Respir Crit Care Med. 1999;160(2):594–599. doi:10.1164/ajrccm.160.2.9811100
  • Rabe KF, Atienza T, Magyar P, Larsson P, Jorup C, Lalloo UG. Effect of budesonide in combination with formoterol for reliever therapy in asthma exacerbations: a randomised controlled, double-blind study. Lancet. 2006;368(9537):744–753. doi:10.1016/S0140-6736(06)69284-2
  • Kuna P, Peters MJ, Manjra AI, et al. Effect of budesonide/formoterol maintenance and reliever therapy on asthma exacerbations. Int J Clin Pract. 2007;61(5):725–736. doi:10.1111/j.1742-1241.2007.01338.x
  • Bousquet J, Boulet LP, Peters MJ, et al. Budesonide/formoterol for maintenance and relief in uncontrolled asthma vs. high-dose salmeterol/fluticasone. Respir Med. 2007;101(12):2437–2446. doi:10.1016/j.rmed.2007.07.014
  • Lazarus SC, Krishnan JA, King TS, et al. Mometasone or tiotropium in mild asthma with a low sputum eosinophil level. N Engl JMed. 2019;380(21):2009–2019. doi:10.1056/NEJMoa1814917
  • Leach C, Colice GL, Luskin A. Particle size of inhaled corticosteroids: does it matter? J Allerg Clin Immunol. 2009;124(6):S88–S93. doi:10.1016/j.jaci.2009.09.050
  • Bacsi A, Choudhury BK, Dharajiya N, Sur S, Boldogh I. Subpollen particles: carriers of allergenic proteins and oxidases. J Allergy Clin Immunol. 2006;118(4):844–850. doi:10.1016/j.jaci.2006.07.006
  • Taylor PE, Flagan RC, Valenta R, Glovsky MM. Release of allergens as respirable aerosols: a link between grass pollen and asthma. J Allergy Clin Immunol. 2002;109(1):51–56. doi:10.1067/mai.2002.120759
  • Adcock IM, Gilbey T, Gelder CM, Chung KF, Barnes PJ. Glucocorticoid receptor localization in normal and asthmatic lung. Am J Respir Crit Care Med. 1996;154(3):771–782. doi:10.1164/ajrccm.154.3.8810618
  • Lavorini F, Pedersen S, Usmani OS, et al. Dilemmas, confusion, and misconceptions related to small airways directed therapy. Chest. 2017;151(6):1345–1355. doi:10.1016/j.chest.2016.07.035
  • Theron AJ, Steel HC, Tintinger GR, Gravett CM, Anderson R, Feldman C. Cysteinyl leukotriene receptor-1 antagonists as modulators of innate immune cell function. J Immunol Res. 2014;2014:608930. doi:10.1155/2014/608930
  • Holgate ST, Peters-Golden M, Panettieri RA, Henderson WR. Roles of cysteinyl leukotrienes in airway inflammation, smooth muscle function, and remodeling. J Allergy Clin Immunol. 2003;111(1):S18–S34. doi:10.1067/mai.2003.25
  • Negri J, Early SB, Steinke JW, Borish L. Corticosteroids as inhibitors of cysteinyl leukotriene metabolic and signaling pathways. J Allergy Clin Immunol. 2008;121(5):1232–1237. doi:10.1016/j.jaci.2008.02.007
  • Gyllfors P, Dahlen SE, Kumlin M, Larsson K, Dahlen B. Bronchial responsiveness to leukotriene D4 is resistant to inhaled fluticasone propionate. J Allergy Clin Immunol. 2006;118(1):78–83. doi:10.1016/j.jaci.2006.03.040
  • Miligkos M, Bannuru RR, Alkofide H, Kher SR, Schmid CH, Balk EM. Leukotriene-receptor antagonists versus placebo in the treatment of asthma in adults and adolescents: a systematic review and meta-analysis. Ann Intern Med. 2015;163(10):756–767. doi:10.7326/M15-1059
  • Zhang HP, Jia CE, Lv Y, Gibson PG, Wang G. Montelukast for prevention and treatment of asthma exacerbations in adults: systematic review and meta-analysis. Allergy Asthma Proc. 2014;35(4):278–287. doi:10.2500/aap.2014.35.3745
  • Marcello C, Carlo L. Asthma phenotypes: the intriguing selective intervention with Montelukast. Asthma Res Pract. 2016;2:11. doi:10.1186/s40733-016-0026-6
  • Gibson PG, Yang IA, Upham JW, et al. Effect of azithromycin on asthma exacerbations and quality of life in adults with persistent uncontrolled asthma (AMAZES): a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10095):659–668. doi:10.1016/S0140-6736(17)31281-3
  • Brusselle GG, Vanderstichele C, Jordens P, et al. Azithromycin for prevention of exacerbations in severe asthma (AZISAST): a multicentre randomised double-blind placebo-controlled trial. Thorax. 2013;68(4):322–329. doi:10.1136/thoraxjnl-2012-202698
  • Shukla SD, Taylor SL, Gibson PG, et al. Add-on azithromycin reduces sputum cytokines in non-eosinophilic asthma: an AMAZES substudy. Thorax. 2021;76:733–736. doi:10.1136/thoraxjnl-2020-216331
  • Niessen NM, Gibson PG, Baines KJ, et al. Sputum TNF markers are increased in neutrophilic and severe asthma and are reduced by azithromycin treatment. Allergy. 2021;76:2090–2101. doi:10.1111/all.14768
  • Sadeghdoust M, Mirsadraee M, Aligolighasemabadi F, Khakzad MR, Hashemi Attar A, Naghibi S. Effect of azithromycin on bronchial wall thickness in severe persistent asthma: a double-blind placebo-controlled randomized clinical trial. Respir Med. 2021;185:106494. doi:10.1016/j.rmed.2021.106494
  • Taylor SL, Ivey KL, Gibson PG, Simpson JL, Rogers GB. Airway abundance of Haemophilus influenzae predicts response to azithromycin in adults with persistent uncontrolled asthma. Eur Respir J. 2020;56(4):2000194. doi:10.1183/13993003.00194-2020
  • Virchow JC. Allergen immunotherapy (AIT) in asthma. Semin Immunol. 2019;46:101334. doi:10.1016/j.smim.2019.101334
  • Virchow JC, Backer V, Kuna P, et al. Efficacy of a house dust mite sublingual allergen immunotherapy tablet in adults with allergic asthma: a randomized clinical trial. JAMA. 2016;315(16):1715–1725. doi:10.1001/jama.2016.3964
  • Agache I, Lau S, Akdis CA, et al. EAACI guidelines on allergen immunotherapy: house dust mite-driven allergic asthma. Allergy. 2019;74(5):855–873. doi:10.1111/all.13749
  • Anonymous. Xolair. European Medicines Agency; 2018.
  • Anonymous. Nucala. European Medicines Agency; 2018.
  • Anonymous. Cinqaero. European Medicines Agency; 2018.
  • Anonymous. Fasenra. European Medicines Agency; 2018.
  • Anonymous. Dupixent. European Medicines Agency; 2018.
  • Brown T, Jones T, Gove K, et al. Randomised controlled trials in severe asthma: selection by phenotype or stereotype. Eur Respir J. 2018;52(6):1801444. doi:10.1183/13993003.01444-2018
  • Travers J, Marsh S, Williams M, et al. External validity of randomised controlled trials in asthma: to whom do the results of the trials apply? Thorax. 2007;62(3):219–233. doi:10.1136/thx.2006.066837
  • Shamji MH, Valenta R, Jardetzky T, et al. The role of allergen-specific IgE, IgG and IgA in allergic disease. Allergy. 2021;Epub. doi:10.1111/all.14908
  • Arroyave WD, Rabito FA, Carlson JC. The relationship between a specific IgE level and asthma outcomes: results from the 2005–2006 national health and nutrition examination survey. J Allergy Clin Immunol Pract. 2013;1(5):501–508.
  • Naqvi M, Choudhry S, Tsai H-J, et al. Association between IgE levels and asthma severity among African American, Mexican, and Puerto Rican patients with asthma. J Allerg Clin Immunol. 2007;120(1):137–143. doi:10.1016/j.jaci.2007.02.045
  • Guntern P, Eggel A. Past, present, and future of anti-IgE biologics. Allergy. 2020;75(10):2491–2502. doi:10.1111/all.14308
  • Ledford D, Busse W, Trzaskoma B, et al. A randomized multicenter study evaluating Xolair persistence of response after long-term therapy. J Allerg Clin Immunol. 2017;140(1):162–169.e162. doi:10.1016/j.jaci.2016.08.054
  • Normansell R, Walker S, Milan SJ, Walters EH, Nair P. Omalizumab for asthma in adults and children. Cochrane Database Syst Rev. 2014;1:CD003559. doi:10.1002/14651858.CD003559.pub4
  • Bousquet J, Humbert M, Gibson PG, et al. Real-world effectiveness of omalizumab in severe allergic asthma: a meta-analysis of observational studies. J Allergy Clin Immunol Pract. 20219(7):2702–2714. doi:10.1016/j.jaip.2021.01.011
  • Alhossan A, Lee CS, MacDonald K, Abraham I. “Real-life” effectiveness studies of omalizumab in adult patients with severe allergic asthma: meta-analysis. J Allergy Clin Immunol Pract. 2017;5(5):1362–1370.e1362. doi:10.1016/j.jaip.2017.02.002
  • Berger W, Gupta N, McAlary M, Fowler-Taylor A. Evaluation of long-term safety of the anti-IgE antibody, omalizumab, in children with allergic asthma. Ann Allerg Asthma Immunol. 2003;91(2):182–188. doi:10.1016/S1081-1206(10)62175-8
  • Di Bona D, Fiorino I, Taurino M, et al. Long-term “real-life” safety of omalizumab in patients with severe uncontrolled asthma: a nine-year study. Respir Med. 2017;130:55–60. doi:10.1016/j.rmed.2017.07.013
  • Long A, Rahmaoui A, Rothman KJ, et al. Incidence of malignancy in patients with moderate-to-severe asthma treated with or without omalizumab. J Allerg Clin Immunol. 2014;134(3):560–567.e564. doi:10.1016/j.jaci.2014.02.007
  • Namazy J, Cabana MD, Scheuerle AE, et al. The Xolair Pregnancy Registry (EXPECT): the safety of omalizumab use during pregnancy. J Allerg Clin Immunol. 2015;135(2):407–412. doi:10.1016/j.jaci.2014.08.025
  • Bousquet J, Rabe K, Humbert M, et al. Predicting and evaluating response to omalizumab in patients with severe allergic asthma. Respir Med. 2007;101(7):1483–1492. doi:10.1016/j.rmed.2007.01.011
  • Wahn U, Martin C, Freeman P, Blogg M, Jimenez P. Relationship between pretreatment specific IgE and the response to omalizumab therapy. Allergy. 2009;64(12):1780–1787. doi:10.1111/j.1398-9995.2009.02119.x
  • Casale TB, Chipps BE, Rosén K, et al. Response to omalizumab using patient enrichment criteria from trials of novel biologics in asthma. Allergy. 2018;73(2):490–497. doi:10.1111/all.13302
  • Hanania NA, Wenzel S, Rosén K, et al. Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am J Respir Crit Care Med. 2013;187(8):804–811. doi:10.1164/rccm.201208-1414OC
  • Casale TB, Luskin AT, Busse W, et al. Omalizumab effectiveness by biomarker status in patients with asthma: evidence from PROSPERO, a prospective real-world study. J Allergy Clin Immunol Pract. 2019;7(1):156–164.e151.
  • Humbert M, Taillé C, Mala L, Gros VL, Just J, Molimard M. Omalizumab effectiveness in patients with severe allergic asthma according to blood eosinophil count: the STELLAIR study. Eur Respir J. 2018;51(5):1702523. doi:10.1183/13993003.02523-2017
  • Tajiri T, Matsumoto H, Gon Y, et al. Utility of serum periostin and free IgE levels in evaluating responsiveness to omalizumab in patients with severe asthma. Allergy. 2016;71(10):1472–1479. doi:10.1111/all.12922
  • Tran TN, Zeiger RS, Peters SP, et al. Overlap of atopic, eosinophilic, and TH2-high asthma phenotypes in a general population with current asthma. Ann Allerg Asthma Immunol. 2016;116(1):37–42. doi:10.1016/j.anai.2015.10.027
  • Bagnasco D, Menzella F, Caminati M, et al. Efficacy of mepolizumab in patients with previous omalizumab treatment failure: real-life observation. Allergy. 2019;74(12):2539–2541. doi:10.1111/all.13937
  • Carpagnano GE, Pelaia C, D’Amato M, et al. Switching from omalizumab to mepolizumab: real-life experience from Southern Italy. Ther Adv Respir Dis. 2020;14:1–13. doi:10.1177/1753466620929231
  • Chapman KR, Albers FC, Chipps B, et al. The clinical benefit of mepolizumab replacing omalizumab in uncontrolled severe eosinophilic asthma. Allergy. 2019;74(9):1716–1726. doi:10.1111/all.13850
  • Farne HA, Wilson A, Powell C, Bax L, Milan SJ. Anti‐IL5 therapies for asthma. Cochrane Database Syst Rev. 2017;9(9):CD010834. doi:10.1002/14651858.CD010834.pub3
  • Park SW, Kim DJ, Chang HS, et al. Association of interleukin-5 and eotaxin with acute exacerbation of asthma. IAA. 2003;131(4):283–290.
  • Kay AB, Phipps S, Robinson DS. A role for eosinophils in airway remodelling in asthma. Trends Immunol. 2004;25(9):477–482. doi:10.1016/j.it.2004.07.006
  • Bel EH, Wenzel SE, Thompson PJ, et al. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N Engl JMed. 2014;371(13):1189–1197. doi:10.1056/NEJMoa1403291
  • 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. doi:10.1056/NEJMoa1403290
  • Bjermer L, Lemiere C, Maspero J, Weiss S, Zangrilli J, Germinaro M. Reslizumab for inadequately controlled asthma with elevated blood eosinophil levels: a randomized phase 3 study. Chest. 2016;150(4):789–798. doi:10.1016/j.chest.2016.03.032
  • Castro M, Zangrilli J, Wechsler ME, et al. Reslizumab for inadequately controlled asthma with elevated blood eosinophil counts: results from two multicentre, parallel, double-blind, randomised, placebo-controlled, phase 3 trials. Lancet Respir Med. 2015;3(5):355–366. doi:10.1016/S2213-2600(15)00042-9
  • Bleecker ER, FitzGerald JM, Chanez P, et al. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting β2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet. 2016;388(10056):2115–2127. doi:10.1016/S0140-6736(16)31324-1
  • FitzGerald JM, Bleecker ER, Nair P, et al. Benralizumab, an anti-interleukin-5 receptor α monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2016;388(10056):2128–2141. doi:10.1016/S0140-6736(16)31322-8
  • Nair P, Wenzel S, Rabe KF, et al. Oral glucocorticoid–sparing effect of benralizumab in severe asthma. N Engl JMed. 2017;376(25):2448–2458. doi:10.1056/NEJMoa1703501
  • Bagnasco D, Caminati M, Menzella F, et al. One year of mepolizumab. Efficacy and safety in real-life in Italy. Pulm Pharmacol Ther. 2019;58(May):101836. doi:10.1016/j.pupt.2019.101836
  • Harvey ES, Langton D, Katelaris C, et al. Mepolizumab effectiveness and identification of super-responders in severe asthma. Eur Respir J. 2020;55(5):1902420. doi:10.1183/13993003.02420-2019
  • Ibrahim H, O’Sullivan R, Casey D, et al. The effectiveness of Reslizumab in severe asthma treatment: a real-world experience. Respir Res. 2019;20(1):289. doi:10.1186/s12931-019-1251-3
  • Kavanagh JE, D’ Ancona G, Elstad M, et al. Real-world effectiveness and the characteristics of a “super-responder” to mepolizumab in severe eosinophilic asthma. Chest. 2020;158(2):491–500. doi:10.1016/j.chest.2020.03.042
  • Kavanagh JE, Hearn AP, D’ Ancona G, et al. Benralizumab after sub-optimal response to mepolizumab in severe eosinophilic asthma. Allergy. 2021;76(6):1890–1893. doi:10.1111/all.14693
  • Menzella F, Bonavia M, Bonini M, et al. Real-world experience with benralizumab in patients with severe eosinophilic asthma: a case series. JAA. 2021;14:149–161. doi:10.2147/JAA.S295676
  • Padilla-Galo A, Levy-Abitbol R, Olveira C, et al. Real-life experience with benralizumab during 6 months. BMC Pulm Med. 2020;20(1):184. doi:10.1186/s12890-020-01220-9
  • Pelaia C, Busceti MT, Vatrella A, et al. Real-life rapidity of benralizumab effects in patients with severe allergic eosinophilic asthma: assessment of blood eosinophils, symptom control, lung function and oral corticosteroid intake after the first drug dose. Pulm Pharmacol Ther. 2019;58:101830. doi:10.1016/j.pupt.2019.101830
  • Pelaia C, Crimi C, Pelaia G, et al. Real-life evaluation of mepolizumab efficacy in patients with severe eosinophilic asthma, according to atopic trait and allergic phenotype. Clin Exp Allerg. 2020;50(7):780–788. doi:10.1111/cea.13613
  • Schleich F, Graff S, Nekoee H, et al. Real-world experience with mepolizumab: does it deliver what it has promised? Clin Exp Allerg. 2020;50(6):687–695. doi:10.1111/cea.13601
  • Wechsler ME, Akuthota P, Jayne D, et al. Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis. N Engl JMed. 2017;376(20):1921–1932. doi:10.1056/NEJMoa1702079
  • Mukherjee M, Bakakos P, Loukides S. New paradigm in asthma management: switching between biologics! Allergy. 2020;75(4):743–745. doi:10.1111/all.14038
  • Pérez de Llano LA, Cosío BG, Domingo C, et al. Efficacy and safety of reslizumab in patients with severe asthma with inadequate response to omalizumab: a multicenter, open-label pilot study. J Allergy Clin Immunol. 2019;7(7):2277–2283.e2272.
  • Burke H, Davis J, Evans S, Flower L, Tan A, Kurukulaaratchy RJ. A multidisciplinary team case management approach reduces the burden of frequent asthma admissions. ERJ Open Res. 2016;2(3):00039–2016. doi:10.1183/23120541.00039-2016
  • 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. 2017;197(1):38–46. doi:10.1164/rccm.201707-1323OC
  • Manetz S, Maric I, Brown T, et al. Successful pregnancy in the setting of eosinophil depletion by benralizumab. J Allergy Clin Immunol Pract. 2021;9(3):1405–1407.e1403.
  • Saco T, Tabatabaian F. Breathing for two: a case of severe eosinophilic asthma during pregnancy treated with benralizumab. Ann Allerg Asthma Immunol. 2018;121(5):S92. doi:10.1016/j.anai.2018.09.300
  • Albers FC, Licskai C, Chanez P, et al. Baseline blood eosinophil count as a predictor of treatment response to the licensed dose of mepolizumab in severe eosinophilic asthma. Respir Med. 2019;159:105806. doi:10.1016/j.rmed.2019.105806
  • Busse W, Chupp G, Nagase H, et al. Anti–IL-5 treatments in patients with severe asthma by blood eosinophil thresholds: indirect treatment comparison. J Allerg Clin Immunol. 2019;143(1):190–200.e120. doi:10.1016/j.jaci.2018.08.031
  • 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. doi:10.1016/S2213-2600(16)30031-5
  • Kavanagh JE, Hearn AP, Dhariwal J, et al. Real world effectiveness of benralizumab in severe eosinophilic asthma. Chest. 2020;158:491–500.
  • Bagnasco D, Massolo A, Bonavia M, et al. The importance of being not significant: blood eosinophils and clinical responses do not correlate in severe asthma patients treated with mepolizumab in real life. Allergy. 2020;75(6):1460–1463. doi:10.1111/all.14135
  • Drick N, Seeliger B, Welte T, Fuge J, Suhling H. Anti-IL-5 therapy in patients with severe eosinophilic asthma - clinical efficacy and possible criteria for treatment response. BMC Pulm Med. 2018;18(1):119. doi:10.1186/s12890-018-0689-2
  • Agache I, Beltran J, Akdis C, et al. Efficacy and safety of treatment with biologicals (benralizumab, dupilumab, mepolizumab, omalizumab and reslizumab) for severe eosinophilic asthma. A systematic review for the EAACI guidelines - recommendations on the use of biologicals in severe asthma. Allergy. 2020;75(5):1023–1042. doi:10.1111/all.14221
  • Rabe KF, Nair P, Brusselle G, et al. Efficacy and safety of dupilumab in glucocorticoid-dependent severe asthma. N Engl JMed. 2018;378:2475–2485. doi:10.1056/NEJMoa1804093
  • Wenzel S, Castro M, Corren J, et al. Dupilumab efficacy and safety in adults with uncontrolled persistent asthma despite use of medium-to-high-dose inhaled corticosteroids plus a long-acting β2 agonist: a randomised double-blind placebo-controlled pivotal phase 2b dose-ranging trial. Lancet. 2016;388(10039):31–44. doi:10.1016/S0140-6736(16)30307-5
  • Corren J, Castro M, O’Riordan T, et al. Dupilumab efficacy in patients with uncontrolled, moderate-to-severe allergic asthma. J Allergy Clin Immunol Pract. 2020;8(2):516–526.
  • Dupin C, Belhadi D, Guilleminault L, et al. Effectiveness and safety of dupilumab for the treatment of severe asthma in a real-life French multi-centre adult cohort. Clin Exp Allerg. 2020;50(7):789–798. doi:10.1111/cea.13614
  • Campisi R, Crimi C, Nolasco S, et al. Real-world experience with dupilumab in severe asthma: one-year data from an Italian named patient program. JAA. 2021;14:575–583. doi:10.2147/JAA.S312123
  • Nowsheen S, Darveaux JI. Real-world efficacy and safety of dupilumab use in the treatment of asthma. Ann Allerg Asthma Immunol. 2021;127:147–149. doi:10.1016/j.anai.2021.04.011
  • Renner A, Marth K, Patocka K, Idzko M, Pohl W. Dupilumab rapidly improves asthma control in predominantly anti-IL5/IL5R pretreated Austrian real-life severe asthmatics. Immun Inflam Dis. 2021;9(3):624–627. doi:10.1002/iid3.434
  • Mümmler C, Munker D, Barnikel M, et al. Dupilumab improves asthma control and lung function in patients with insufficient outcome during previous antibody therapy. J Allergy Clin Immunol. 2021;9(3):1177–1185.e1174.
  • Bosma AL, Gerbens LA, Middelkamp-Hup MA, Spuls PI. Paternal and maternal use of dupilumab in patients with atopic dermatitis: a case series. Clin Exp Dermatol. 202146(6):1089–1092. doi:10.1111/ced.14725
  • Kage P, Simon JC, Treudler R. A case of atopic eczema treated safely with dupilumab during pregnancy and lactation. J Eur Acad Dermatol Venereol. 2020;34(6):e256–e257. doi:10.1111/jdv.16235
  • Mian M, Dunlap R, Simpson E. Dupilumab for the treatment of severe atopic dermatitis in a pregnant patient: a case report. JAAD Case Rep. 2020;6(10):1051–1052. doi:10.1016/j.jdcr.2020.08.001
  • Regeneron P. Registry of Asthma Patients Initiating DUPIXENT® (RAPID). 2021. 05 June, 2021. NCT04287621.
  • Guntur VP, Manka LA, Denson JL, et al. Benralizumab as a steroid-sparing treatment option in eosinophilic granulomatosis with polyangiitis. J Allergy Clin Immunol. 2021;9(3):1186–1193.e1181.
  • Han JK, Bachert C, Fokkens W, et al. Mepolizumab for chronic rhinosinusitis with nasal polyps (SYNAPSE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2021;Epub. doi:10.1016/S2213-2600(21)00097-7
  • Takabayashi T, Asaka D, Okamoto Y, et al. A phase II, multicenter, randomized, placebo-controlled study of benralizumab, a humanized anti-IL-5R alpha monoclonal antibody, in patients with eosinophilic chronic rhinosinusitis. Am J Rhinol Allerg. 2021;Epub. doi:10.1177/19458924211009429
  • Tversky J, Lane AP, Azar A. Benralizumab effect on severe chronic rhinosinusitis with nasal polyps (CRSwNP): a randomized double-blind placebo-controlled trial. Clin Exp Allerg. 2021;51(6):836–844. doi:10.1111/cea.13852
  • Assa’ad AH, Gupta SK, Collins MH, et al. An antibody against IL-5 reduces numbers of esophageal intraepithelial eosinophils in children with eosinophilic esophagitis. Gastroenterology. 2011;141(5):1593–1604. doi:10.1053/j.gastro.2011.07.044
  • Menzies-Gow A, Corren J, Bourdin A, et al. Tezepelumab in adults and adolescents with severe, uncontrolled asthma. N Engl JMed. 2021;384(19):1800–1809. doi:10.1056/NEJMoa2034975
  • Peters MC, Wenzel SE. Intersection of biology and therapeutics: type 2 targeted therapeutics for adult asthma. Lancet. 2020;395(10221):371–383. doi:10.1016/S0140-6736(19)33005-3
  • Ziegler SF, Roan F, Bell BD, Stoklasek TA, Kitajima M, Han H. The biology of thymic stromal lymphopoietin (TSLP). Adv Pharmacol. 2013;66:129–155.
  • Porsbjerg CM, Sverrild A, Lloyd CM, Menzies-Gow AN, Bel EH. Anti-alarmins in asthma: targeting the airway epithelium with next-generation biologics. Eur Respir J. 2020;56(5):2000260. doi:10.1183/13993003.00260-2020
  • Brightling C, Kulkarni S, Lambrecht BN, Sandham D, Weiss M, Altman P. The pharmacology of the prostaglandin D2 receptor 2 (DP2) receptor antagonist, fevipiprant. Pulm Pharmacol Ther. 2021;68:102030. doi:10.1016/j.pupt.2021.102030
  • Prussin C, Panettieri RA, Bozik ME, Archibald DG, Mather JL, Siddiqui S. Oral dexpramipexole efficacy in lowering blood eosinophils in patients with moderate to severe uncontrolled eosinophilic asthma: study design and baseline data from the AS201 phase 2 trial. Am J Respir Crit Care Med. 2021;203:A1359.