770
Views
0
CrossRef citations to date
0
Altmetric
Review

Mild asthma is not mild: risk factors and predictive biomarkers for severe acute exacerbations and progression in mild asthma

, , , & ORCID Icon
Pages 1261-1271 | Received 28 Oct 2023, Accepted 01 Feb 2024, Published online: 06 Feb 2024

References

  • Disease GBD, Injury I, Prevalence C. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the global burden of disease study. 2016. Lancet. 2017;390(10100):1211–1259. doi: 10.1016/S0140-6736(17)32154-2
  • Huang K, Yang T, Xu J, et al. Prevalence, risk factors, and management of asthma in China: a national cross-sectional study. Lancet. 2019;394(10196):407–418. doi: 10.1016/S0140-6736(19)31147-X
  • Hekking P-P, Wener RR, Amelink M, et al. The prevalence of severe refractory asthma. J Allergy Clin Immunol. 2015;135(4):896–902. doi: 10.1016/j.jaci.2014.08.042
  • 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
  • Fitzgerald JM, Barnes PJ, Chipps BE, et al. The burden of exacerbations in mild asthma: a systematic review. ERJ Open Res. 2020;6(3):00359–2019. doi: 10.1183/23120541.00359-2019
  • Mulgirigama A, Barnes N, Fletcher M, et al. A review of the burden and management of mild asthma in adults — implications for clinical practice. Respir med. 2019;152:97–104. doi: 10.1016/j.rmed.2019.04.024
  • Tang W, Sun L, Fizgerald JM. A paradigm shift in the treatment of mild asthma? J Thoracic Dis. 2018;10(10):5655–5658. doi: 10.21037/jtd.2018.09.127
  • Cloutier MM, Dixon AE, Krishnan JA, et al. Managing asthma in adolescents and adults: 2020 asthma guideline update from the national asthma education and prevention program. JAMA. 2020;324(22):2301–2317. doi: 10.1001/jama.2020.21974
  • Ding B, Dibonaventura M, Karlsson N, et al. A cross-sectional assessment of the prevalence and burden of mild asthma in urban China using the 2010, 2012, and 2013 China national health and wellness surveys. J Asthma. 2017;54(6):632–643. doi: 10.1080/02770903.2016.1255750
  • Price D, Fletcher M, Van Der Molen T. Asthma control and management in 8,000 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
  • Colice GL, Ostrom NK, Geller DE, et al. The CHOICE survey: high rates of persistent and uncontrolled asthma in the United States. Ann Allergy Asthma Immunol. 2012;108(3):157–162. doi: 10.1016/j.anai.2011.12.017
  • Ding B, Small M. Disease burden of mild asthma in China. Respirology. 2018;23(4):369–377. doi: 10.1111/resp.13189
  • Mohan A, Lugogo NL, Hanania NA, et al. Questions in mild asthma: an official American thoracic society research statement. Am J Respir Crit Care Med. 2023;207(11):e77–e96. doi: 10.1164/rccm.202304-0642ST
  • Ernst P, Cai B, Blais L, et al. The early course of newly diagnosed asthma. Am J Med. 2002;112(1):44–48. doi: 10.1016/s0002-9343(01)01033-6
  • Chen W, Fitzgerald JM, Lynd LD, et al. Long-term trajectories of mild asthma in adulthood and risk factors of progression. J Allergy Clin Immunol Pract. 2018;6(6):2024–2032.e5. doi: 10.1016/j.jaip.2018.04.027
  • Anderson GP. Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet. 2008;372(9643):1107–1119. doi: 10.1016/S0140-6736(08)61452-X
  • Moffatt MF, Gut IG, Demenais F, et al. A large-scale, consortium-based genomewide association study of asthma. N Engl J Med. 2010;363(13):1211–1221. doi: 10.1056/NEJMoa0906312
  • Wang J, Zhou Y, Zhang H, et al. Pathogenesis of allergic diseases and implications for therapeutic interventions. Signal Transduct Target Ther. 2023;8(1):138. doi: 10.1038/s41392-023-01344-4
  • Dong Z, Ma Y, Zhou H, et al. Integrated genomics analysis highlights important SNPs and genes implicated in moderate-to-severe asthma based on GWAS and eQTL datasets. BMC Pulm Med. 2020;20(1):270. doi: 10.1186/s12890-020-01303-7
  • Belsky DW, Sears MR, Hancox RJ, et al. Polygenic risk and the development and course of asthma: an analysis of data from a four-decade longitudinal study. Lancet Respir Med. 2013;1(6):453–461. doi: 10.1016/S2213-2600(13)70101-2
  • Schoettler N, Ober C. Genetic architecture of moderate-to-severe asthma mirrors that of mild asthma. J Allergy Clin Immunol. 2019;144(6):1521–1523. doi: 10.1016/j.jaci.2019.09.003
  • Hinds DA, Mcmahon G, Kiefer AK, et al. A genome-wide association meta-analysis of self-reported allergy identifies shared and allergy-specific susceptibility loci. Nat Genet. 2013;45(8):907–911. doi: 10.1038/ng.2686
  • Gautam Y, Johansson E, Mersha TB. Multi-omics profiling approach to asthma: an evolving paradigm. J Pers Med. 2022;12(1):66. doi: 10.3390/jpm12010066
  • Herrera-Luis E, Forno E, Celedón JC, et al. Asthma exacerbations: the genes behind the scenes. J Investig Allergol Clin. 2023;33(2):76–94. doi: 10.18176/jiaci.0878
  • Vignola AM, Chanez P, Campbell AM, et al. Airway inflammation in mild intermittent and in persistent asthma. Am J Respir Crit Care Med. 1998;157(2):403–409. doi: 10.1164/ajrccm.157.2.96-08040
  • Laprise C, Laviolette M, Boutet M, et al. Asymptomatic airway hyperresponsiveness: relationships with airway inflammation and remodelling. Eur Respir J. 1999;14(1):63–73. doi: 10.1034/j.1399-3003.1999.14a12.x
  • Spallarossa D, Battistini E, Silvestri M, et al. Steroid-naive adolescents with mild intermittent allergic asthma have airway hyperresponsiveness and elevated exhaled nitric oxide levels. J Asthma. 2003;40(3):301–310. doi: 10.1081/jas-120018629
  • Fehrenbach H, Wagner C, Wegmann M. Airway remodeling in asthma: what really matters. Cell Tissue Res. 2017;367(3):551–569. doi: 10.1007/s00441-016-2566-8
  • Boulet LP, Jobin C, Milot J, et al. Five-year changes in airflow obstruction and airway responsiveness in mild to moderate asthma. Clin Invest Med. 1994;17(5):432–442.
  • Wardlaw AJ, Dunnette S, Gleich GJ, et al. Eosinophils and mast cells in bronchoalveolar lavage in subjects with mild asthma. relationship to bronchial hyperreactivity. Am Rev Respir Dis. 1988;137(1):62–69. doi: 10.1164/ajrccm/137.1.62
  • Boulet L-P. Airway remodeling in asthma: update on mechanisms and therapeutic approaches. Curr Opin Pulm Med. 2018;24(1):56–62. doi: 10.1097/MCP.0000000000000441
  • Postma DS, Rabe KF. The Asthma-COPD overlap syndrome. N Engl J Med. 2015;373(13):1241–1249. doi: 10.1056/NEJMra1411863
  • Kole TM, Vanden Berghe E, Kraft M, et al. Predictors and associations of the persistent airflow limitation phenotype in asthma: a post-hoc analysis of the ATLANTIS study. Lancet Respir Med. 2023;11(1):55–64. doi: 10.1016/S2213-2600(22)00185-0
  • Matsunaga K, Hirano T, Oka A, et al. Progression of irreversible airflow limitation in asthma: correlation with severe exacerbations. J Allergy Clin Immunol Pract. 2015;3(5):759–64.e1. doi: 10.1016/j.jaip.2015.05.005
  • Siroux V, Boudier A, Dolgopoloff M, et al. Forced midexpiratory flow between 25% and 75% of forced vital capacity is associated with long-term persistence of asthma and poor asthma outcomes. J Allergy Clin Immunol. 2016;137(6):759–64.e1. doi: 10.1016/j.jaci.2015.10.029
  • Ban GY, Kim SC, Lee HY, et al. Risk factors predicting severe asthma exacerbations in adult asthmatics: a real-world clinical evidence. Allergy Asthma Immunol Res. 2021;13(3):420–434. doi: 10.4168/aair.2021.13.3.420
  • Vedel-Krogh S, Fallgaard Nielsen S, Lange P, et al. Association of blood eosinophil and blood neutrophil counts with asthma exacerbations in the Copenhagen General Population Study. Clin Chem. 2017;63(4):823–832. doi: 10.1373/clinchem.2016.267450
  • 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
  • Zeiger RS, Schatz M, Li Q, et al. High blood eosinophil count is a risk factor for future asthma exacerbations in adult persistent asthma. J Allergy Clin Immunol Pract. 2014;2(6):741–750. doi: 10.1016/j.jaip.2014.06.005
  • Marc-Malovrh M, Camlek L, Škrgat S, et al. Elevated eosinophils, IL5 and IL8 in induced sputum in asthma patients with accelerated FEV1 decline. Respir med. 2020;162:105875. doi: 10.1016/j.rmed.2020.105875
  • Ciółkowski J, Mazurek H, Hydzik P, et al. Inflammatory markers as exacerbation risk factors after asthma therapy switch from inhaled steroids to montelukast. Pulm Pharmacol Ther. 2016;39:7–13. doi: 10.1016/j.pupt.2016.05.002
  • Tillie-Leblond I, Montani D, Crestani B, et al. Relation between inflammation and symptoms in asthma. Allergy. 2009;64(3):354–367. doi: 10.1111/j.1398-9995.2009.01971.x
  • Bel EH, Wenzel SE, Thompson PJ, et al. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N Engl J Med. 2014;371(13):1189–1197. doi: 10.1056/NEJMoa1403291
  • 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
  • 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
  • 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
  • Flood-Page PT, Menzies-Gow AN, Kay AB, et al. 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
  • Leckie MJ, Ten Brinke A, Khan J, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000;356(9248):2144–2148. doi: 10.1016/s0140-6736(00)03496-6
  • Sabogal Piñeros YS, Bal SM, De Pol MA V, et al. Anti-IL-5 in mild asthma alters rhinovirus-induced macrophage, B-Cell, and neutrophil responses (MATERIAL). A placebo-controlled, double-blind study. Am J Respir Crit Care Med. 2019;199(4):508–517. doi: 10.1164/rccm.201803-0461OC
  • Vähätalo I, Lehtimäki L, Tuomisto LE, et al. Long-term use of short-acting β2-agonists in patients with adult-onset asthma. J Allergy Clin Immunol Pract. 2022;10(8):2074–2083.e7. doi: 10.1016/j.jaip.2022.03.027
  • Amin S, Soliman M, Mcivor A, et al. Usage patterns of short-acting β2-agonists and inhaled corticosteroids in asthma: a targeted literature review. J Allergy Clin Immunol Pract. 2020;8(8):2556–2564.e8. doi: 10.1016/j.jaip.2020.03.013
  • Nwaru BI, Ekström M, Hasvold P, et al. Overuse of short-acting β2-agonists in asthma is associated with increased risk of exacerbation and mortality: a nationwide cohort study of the global SABINA programme. Eur Respir J. 2020;55(4):1901872. doi: 10.1183/13993003.01872-2019
  • Bateman ED, Price DB, Wang H-C, et al. Short-acting β2-agonist prescriptions are associated with poor clinical outcomes of asthma: the multi-country, cross-sectional SABINA III study. Eur Respir J. 2022;59(5):2101402. doi: 10.1183/13993003.01402-2021
  • Pauwels RA, Pedersen S, Busse WW, et al. Early intervention with budesonide in mild persistent asthma: a randomised, double-blind trial. Lancet. 2003;361(9363):1071–1076. doi: 10.1016/S0140-6736(03)12891-7
  • O’byrne PM, Barnes PJ, Rodriguez-Roisin R, et al. Low dose inhaled budesonide and formoterol in mild persistent asthma: the OPTIMA randomized trial. Am J Respir Crit Care Med. 2001;164(8 Pt 1):1392–1397.
  • Romagnoli M, Caramori G, Braccioni F, et al. Near-fatal asthma phenotype in the ENFUMOSA cohort. Clin Exp Allergy. 2007;37(4):552–557. doi: 10.1111/j.1365-2222.2007.02683.x
  • Hogan MB, Zhao W. High SABA Using and Poor Asthma Control: Blaming Poor Adherence or Missing the Phenotype? J Allergy Clin Immunol Pract. 2022;10(8):2084–2085. doi: 10.1016/j.jaip.2022.05.011
  • Gregoriano C, Dieterle T, Breitenstein A-L, et al. Use and inhalation technique of inhaled medication in patients with asthma and COPD: data from a randomized controlled trial. Respir Res. 2018;19(1):237. doi: 10.1186/s12931-018-0936-3
  • Usmani OS, Lavorini F, Marshall J, et al. Critical inhaler errors in asthma and COPD: a systematic review of impact on health outcomes. Respir Res. 2018;19(1):10. doi: 10.1186/s12931-017-0710-y
  • Aldridge RE, Hancox RJ, Robin Taylor D, et al. Effects of terbutaline and budesonide on sputum cells and bronchial hyperresponsiveness in asthma. Am J Respir Crit Care Med. 2000;161(5):1459–1464. doi: 10.1164/ajrccm.161.5.9906052
  • Hancox RJ, Cowan JO, Flannery EM, et al. Bronchodilator tolerance and rebound bronchoconstriction during regular inhaled beta-agonist treatment. Respir med. 2000;94(8):767–771. doi: 10.1053/rmed.2000.0820
  • Nannini LJ. It is time to end our love affair with short-acting β2-agonists in asthma? yes. ERJ Open Res. 2022;8(4):00353–2022. doi: 10.1183/23120541.00353-2022
  • Kang H-R, Song HJ, Nam JH, et al. Risk factors of asthma exacerbation based on asthma severity: a nationwide population-based observational study in South Korea. BMJ Open. 2018;8(3):e020825. doi: 10.1136/bmjopen-2017-020825
  • Carroll CL, Uygungil B, Zucker AR, et al. Identifying an at-risk population of children with recurrent near-fatal asthma exacerbations. J Asthma. 2010;47(4):460–464. doi: 10.3109/02770903.2010.481344
  • Sekiya K, Nakatani E, Fukutomi Y, et al. Severe or life-threatening asthma exacerbation: patient heterogeneity identified by cluster analysis. Clin Exp Allergy. 2016;46(8):1043–1055. doi: 10.1111/cea.12738
  • Woo S-D, Park HS, Jang J-H, et al. Biomarkers for predicting type 2-high and uncontrolled asthma in real-world practice. Ann Allergy Asthma Immunol. 2023;131(2):209–216.e2. doi: 10.1016/j.anai.2023.05.011
  • Buhl R, Korn S, Menzies-Gow A, et al. Prospective, single-arm, longitudinal study of biomarkers in real-world patients with severe asthma. J Allergy Clin Immunol Pract. 2020;8(8):2630–2639.e6. doi: 10.1016/j.jaip.2020.03.038
  • Kraft M, Brusselle G, Fitzgerald JM, et al. Patient characteristics, biomarkers and exacerbation risk in severe, uncontrolled asthma. Eur Respir J. 2021;58(6):2100413. doi: 10.1183/13993003.00413-2021
  • Pavord ID, Holliday M, Reddel HK, et al. Predictive value of blood eosinophils and exhaled nitric oxide in adults with mild asthma: a prespecified subgroup analysis of an open-label, parallel-group, randomised controlled trial. Lancet Respir Med. 2020;8(7):671–680. doi: 10.1016/S2213-2600(20)30053-9
  • Hanibuchi M, Mitsuhashi A, Kajimoto T, et al. Clinical significance of fractional exhaled nitric oxide and periostin as potential markers to assess therapeutic efficacy in patients with cough variant asthma. Respir Investig. 2023;61(1):16–22. doi: 10.1016/j.resinv.2022.10.006
  • Mansur AH, Srivastava S, Sahal A. Disconnect of type 2 biomarkers in severe asthma; dominated by FeNO as a predictor of exacerbations and periostin as predictor of reduced lung function. Respir med. 2018;143:31–38. doi: 10.1016/j.rmed.2018.08.005
  • Peters MC, Mauger D, Ross KR, et al. Evidence for exacerbation-prone asthma and predictive biomarkers of exacerbation frequency. Am J Respir Crit Care Med. 2020;202(7):973–982. doi: 10.1164/rccm.201909-1813OC
  • Zhou T, Huang X, Ma J, et al. Association of plasma soluble CD14 level with asthma severity in adults: a case control study in China. Respir Res. 2019;20(1):19. doi: 10.1186/s12931-019-0987-0
  • Sigari N, Jalili A, Mahdawi L, et al. Soluble CD93 as a novel biomarker in asthma exacerbation. Allergy Asthma Immunol Res. 2016;8(5):461–465. doi: 10.4168/aair.2016.8.5.461
  • Ip WK, Wong CK, Leung TF, et al. Plasma concentrations of soluble CTLA-4, CD28, CD80 and CD86 costimulatory molecules reflect disease severity of acute asthma in children. Pediatr Pulmonol. 2006;41(7):674–682. doi: 10.1002/ppul.20432
  • Jones AC, Leffler J, Laing IA, et al. LPS binding protein and activation signatures are upregulated during asthma exacerbations in children. Respir Res. 2023;24(1):184. doi: 10.1186/s12931-023-02478-3
  • Watanabe M, Nakamoto K, Inui T, et al. Serum sST2 levels predict severe exacerbation of asthma. Respir Res. 2018;19(1):169. doi: 10.1186/s12931-018-0872-2
  • Fricker M, Gibson PG, Powell H, et al. A sputum 6-gene signature predicts future exacerbations of poorly controlled asthma. J Allergy Clin Immunol. 2019;144(1):51–60.e11. doi: 10.1016/j.jaci.2018.12.1020
  • Suzuki Y, Saito J, Kikuchi M, et al. Sputum-to-serum hydrogen sulphide ratio as a novel biomarker of predicting future risks of asthma exacerbation. Clin Exp Allergy. 2018;48(9):1155–1163. doi: 10.1111/cea.13173
  • Choi S, Sohn K-H, Jung J-W, et al. Lung virome: new potential biomarkers for asthma severity and exacerbation. J Allergy Clin Immunol. 2021;148(4):1007–1015.e9. doi: 10.1016/j.jaci.2021.03.017
  • Kraft M, Richardson M, Hallmark B, et al. The role of small airway dysfunction in asthma control and exacerbations: a longitudinal, observational analysis using data from the ATLANTIS study. Lancet Respir Med. 2022;10(7):661–668. doi: 10.1016/S2213-2600(21)00536-1
  • Kim MS, Cho YJ, Moon HB, et al. Factors for poor prognosis of near-fatal asthma after recovery from a life-threatening asthma attack. Korean J Intern Med. 2008;23(4):170–175. doi: 10.3904/kjim.2008.23.4.170
  • Bateman ED, Reddel HK, O’byrne PM, et al. As-Needed Budesonide-Formoterol versus Maintenance Budesonide in Mild Asthma. N Engl J Med. 2018;378(20):1877–1887. doi: 10.1056/NEJMoa1715275
  • Domingo C, Rello J, Sogo A. As-needed ICS-LABA in mild asthma: what does the evidence say? Drugs. 2019;79(16):1729–1737. doi: 10.1007/s40265-019-01202-0
  • Daley-Yates P, Singh D, Igea JM, et al. Assessing the effects of changing patterns of inhaled corticosteroid dosing and adherence with fluticasone furoate and budesonide on asthma management. Adv Ther. 2023;40(9):4042–4059. doi: 10.1007/s12325-023-02585-z
  • O’byrne PM, Fitzgerald JM, Bateman ED, et al. Effect of a single day of increased as-needed budesonide-formoterol use on short-term risk of severe exacerbations in patients with mild asthma: a post-hoc analysis of the SYGMA 1 study. Lancet Respir Med. 2021;9(2):149–158. doi: 10.1016/S2213-2600(20)30416-1
  • Reddel HK, O’byrne PM, Fitzgerald JM, et al. Efficacy and safety of as-needed budesonide-formoterol in adolescents with mild asthma. J Allergy Clin Immunol Pract. 2021;9(8):3069–3077.e6. doi: 10.1016/j.jaip.2021.04.016
  • 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
  • Tong X, Liu T, Li Z, et al. Is it really feasible to use budesonide-formoterol as needed for mild persistent asthma? A systematic review and meta-analysis. Front Pharmacol. 2021;12:644629. doi: 10.3389/fphar.2021.644629
  • Bateman ED, O’byrne PM, Fitzgerald JM, et al. Positioning As-needed budesonide-formoterol for mild asthma: effect of prestudy treatment in pooled analysis of SYGMA 1 and 2. Ann Am Thorac Soc. 2021;18(12):2007–2017. doi: 10.1513/AnnalsATS.202011-1386OC
  • Fitzgerald JM, O’byrne PM, Bateman ED, et al. Safety of As-needed budesonide-formoterol in mild asthma: data from the two phase III SYGMA studie. Drug Saf. 2021;44(4):467–478. doi: 10.1007/s40264-020-01041-z
  • Fitzgerald JM, Arnetorp S, Smare C, et al. The cost-effectiveness of as-needed budesonide/formoterol versus low-dose inhaled corticosteroid maintenance therapy in patients with mild asthma in the UK. Respir med. 2020;171:106079. doi: 10.1016/j.rmed.2020.106079
  • Buendía JA, Patiño DG. Cost-utility of as-needed ICS-formoterol versus to maintenance ICS in mild to moderate persistent asthma. BMC Pulm Med. 2021;21(1):397. doi: 10.1186/s12890-021-01775-1
  • Bateman ED, Hurd SS, Barnes PJ, et al. Global strategy for asthma management and prevention: GINA executive summary. Eur Respir J. 2008;31(1):143–178. doi: 10.1183/09031936.00138707