1932

Abstract

The presentation, pathobiology, and prognosis of asthma are highly heterogeneous and challenging for clinicians to diagnose and treat. In addition to the adaptive immune response that underlies allergic inflammation, innate immune mechanisms are increasingly recognized to be critical mediators of the eosinophilic airway inflammation present in most patients with asthma. Efforts to classify patients by severity and immune response have identified a number of different clinical and immune phenotypes, indicating that the innate and adaptive immune responses are differentially active among patients with the disease. Advances in the detection of these subgroups using clinical characteristics and biomarkers have led to the successful development of targeted biologics. This has moved us to a more personalized approach to managing asthma. Here we review the emerging endotypes of asthma and the biologics that have been developed to treat them.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-041818-020630
2020-01-27
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/med/71/1/annurev-med-041818-020630.html?itemId=/content/journals/10.1146/annurev-med-041818-020630&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Moorman JE, Zahran H, Truman BI, Molla MT 2011. Current asthma prevalence—United States, 2006–2008. MMWR Suppl 60:184–86
    [Google Scholar]
  2. 2. 
    Cent. Dis. Control Prev 2011. Vital signs: asthma prevalence, disease characteristics, and self-management education: United States, 2001–2009. Morb. Mortal. Wkly. Rep 60:17547–52
    [Google Scholar]
  3. 3. 
    Moore WC, Peters SP. 2006. Severe asthma: an overview. J. Allergy Clin. Immunol. 117:3487–94
    [Google Scholar]
  4. 4. 
    Cohn L, Elias JA, Chupp GL 2004. Asthma: mechanisms of disease persistence and progression. Annu. Rev. Immunol. 22:789–815
    [Google Scholar]
  5. 5. 
    Chung KF, Wenzel S. 2014. From the authors: International European Respiratory Society/American Thoracic Society guidelines on severe asthma. Eur. Respir. J. 44:51378–79
    [Google Scholar]
  6. 6. 
    Childhood Asthma Manag. Program Res. Group 2000. Long-term effects of budesonide or nedocromil in children with asthma. N. Engl. J. Med 343:151054–63
    [Google Scholar]
  7. 7. 
    Wenzel S. 2012. Severe asthma: from characteristics to phenotypes to endotypes. Clin. Exp. Allergy 42:5650–58
    [Google Scholar]
  8. 8. 
    Kaur R, Chupp G. 2019. Phenotypes and endotypes of adult asthma: moving toward precision medicine. J. Allergy Clin. Immunol. 144:11–12
    [Google Scholar]
  9. 9. 
    Ogawa Y, Calhoun WJ. 2010. Phenotypic characterization of severe asthma. Curr. Opin. Pulm. Med. 16:148–54
    [Google Scholar]
  10. 10. 
    Yan X, Chu JH, Gomez J et al. 2016. Noninvasive analysis of the sputum transcriptome discriminates clinical phenotypes of asthma. Ann. Am. Thorac. Soc. 13:Suppl. 1S104–5
    [Google Scholar]
  11. 11. 
    Moore WC, Meyers DA, Wenzel SE et al. 2010. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am. J. Respir. Crit. Care Med. 181:4315–23
    [Google Scholar]
  12. 12. 
    Chupp GL, Lee CG, Jarjour N et al. 2007. A chitinase-like protein in the lung and circulation of patients with severe asthma. N. Engl. J. Med. 357:202016–27
    [Google Scholar]
  13. 13. 
    Chung KF, Wenzel SE, Brozek JL et al. 2014. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur. Respir. J. 43:2343–73
    [Google Scholar]
  14. 14. 
    Boulet LP, Reddel H, Bateman E et al. 2019. The Global Initiative for Asthma (GINA): 25 years later. Eur. Respir. J. 54:1900598
    [Google Scholar]
  15. 15. 
    Lambrecht BN, Hammad H, Fahy JV 2019. The cytokines of asthma. Immunity 50:4975–91
    [Google Scholar]
  16. 16. 
    Zhu Z, Lee CG, Zheng T et al. 2001. Airway inflammation and remodeling in asthma—lessons from interleukin 11 and interleukin 13 transgenic mice. Am. J. Respir. Crit. Care Med. 164:10S67–S70
    [Google Scholar]
  17. 17. 
    Zhu Z, Homer RJ, Wang Z et al. 1999. Pulmonary expression of interleukin-13 causes inflammation, mucus hypersecretion, subepithelial fibrosis, physiologic abnormalities, and eotaxin production. J. Clin. Investig. 103:6779–88
    [Google Scholar]
  18. 18. 
    Price DB, Rigazio A, Campbell JD et al. 2015. Blood eosinophil count and prospective annual asthma disease burden: a UK cohort study. Lancet Respir. Med. 3:11849–58
    [Google Scholar]
  19. 19. 
    Price D, Wilson AM, Chisholm A et al. 2016. Predicting frequent asthma exacerbations using blood eosinophil count and other patient data routinely available in clinical practice. J. Asthma Allergy 9:1–12
    [Google Scholar]
  20. 20. 
    Tran TN, Khatry DB, Ke X et al. 2014. High blood eosinophil count is associated with more frequent asthma attacks in asthma patients. Ann. Allergy Asthma Immunol. 113:119–24
    [Google Scholar]
  21. 21. 
    Corren J, Parnes JR, Wang L et al. 2017. Tezepelumab in adults with uncontrolled asthma. N. Engl. J. Med. 377:10936–46
    [Google Scholar]
  22. 22. 
    Green RH, Brightling CE, McKenna S et al. 2002. Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet 360:93471715–21
    [Google Scholar]
  23. 23. 
    Buhl R, Humbert M, Bjermer L et al. 2017. Severe eosinophilic asthma: a roadmap to consensus. Eur. Respir. J. 49:51700329
    [Google Scholar]
  24. 24. 
    Nair P, O'Byrne PM. 2016. Measuring eosinophils to make treatment decisions in asthma. Chest 150:3485–87
    [Google Scholar]
  25. 25. 
    Piacentini GL, Bodini A, Costella S et al. 2000. Exhaled nitric oxide is reduced after sputum induction in asthmatic children. Pediatr. Pulmonol. 29:6430–33
    [Google Scholar]
  26. 26. 
    Pifferi M, Bush A, Pioggia G et al. 2011. Monitoring asthma control in children with allergies by soft computing of lung function and exhaled nitric oxide. Chest 139:2319–27
    [Google Scholar]
  27. 27. 
    Dweik RA, Sorkness RL, Wenzel S et al. 2010. Use of exhaled nitric oxide measurement to identify a reactive, at-risk phenotype among patients with asthma. Am. J. Respir. Crit. Care Med. 181:101033–41
    [Google Scholar]
  28. 28. 
    Piacentini GL, Bodini A, Costella S et al. 1999. Exhaled nitric oxide and sputum eosinophil markers of inflammation in asthmatic children. Eur. Respir. J. 13:61386–90
    [Google Scholar]
  29. 29. 
    Smith AD, Cowan JO, Brassett KP et al. 2005. Use of exhaled nitric oxide measurements to guide treatment in chronic asthma. N. Engl. J. Med. 352:212163–73
    [Google Scholar]
  30. 30. 
    Casale TB, Chipps BE, Rosen K et al. 2018. Response to omalizumab using patient enrichment criteria from trials of novel biologics in asthma. Allergy 73:2490–97
    [Google Scholar]
  31. 31. 
    Busse W, Corren J, Lanier BQ et al. 2001. Omalizumab, anti-IgE recombinant humanized monoclonal antibody, for the treatment of severe allergic asthma. J. Allergy Clin. Immunol. 108:2184–90
    [Google Scholar]
  32. 32. 
    Hanania NA, Alpan O, Hamilos DL et al. 2011. Omalizumab in severe allergic asthma inadequately controlled with standard therapy: a randomized trial. Ann. Intern. Med. 154:9573–82
    [Google Scholar]
  33. 33. 
    Hanania NA, Wenzel S, Rosen K et al. 2013. Exploring the effects of omalizumab in allergic asthma: an analysis of biomarkers in the EXTRA study. Am. J. Respir. Crit. Care Med. 187:8804–11
    [Google Scholar]
  34. 34. 
    Ortega G, Tongchinsub P, Carr T 2019. Combination biologic therapy for severe persistent asthma. Ann. Allergy Asthma Immunol. 123:3309–11
    [Google Scholar]
  35. 35. 
    Chipps BE, Lanier B, Milgrom H et al. 2017. Omalizumab in children with uncontrolled allergic asthma: review of clinical trial and real-world experience. J. Allergy Clin. Immunol. 139:51431–44
    [Google Scholar]
  36. 36. 
    Wu KCP, Jabbar-Lopez ZK. 2015. Omalizumab, an anti-IgE mAb, receives approval for the treatment of chronic idiopathic/spontaneous urticaria. J. Investig. Dermatol. 135:113–15
    [Google Scholar]
  37. 37. 
    Wang FP, Xiong XF, Liu T et al. 2018. Anti-interleukin 5 therapy for eosinophilic asthma: a meta-analysis of randomized clinical trials. Clin. Rev. Allergy Immunol. 54:2318–30
    [Google Scholar]
  38. 38. 
    Wang FP, Liu T, Lan Z et al. 2016. Efficacy and safety of anti-interleukin-5 therapy in patients with asthma: a systematic review and meta-analysis. PLOS ONE 11:11e0166833
    [Google Scholar]
  39. 39. 
    Ortega HG, Liu MC, Pavord ID et al. 2014. Mepolizumab treatment in patients with severe eosinophilic asthma. N. Engl. J. Med. 371:131198–207
    [Google Scholar]
  40. 40. 
    Haldar P, Brightling CE, Hargadon B et al. 2009. Mepolizumab and exacerbations of refractory eosinophilic asthma. N. Engl. J. Med. 360:10973–84
    [Google Scholar]
  41. 41. 
    Nair P, Pizzichini MM, Kjarsgaard M et al. 2009. Mepolizumab for prednisone-dependent asthma with sputum eosinophilia. N. Engl. J. Med. 360:10985–93
    [Google Scholar]
  42. 42. 
    Flood-Page P, Swenson C, Faiferman I et al. 2007. A study to evaluate safety and efficacy of mepolizumab in patients with moderate persistent asthma. Am. J. Respir. Crit. Care Med. 176:111062–71
    [Google Scholar]
  43. 43. 
    Pavord ID, Korn S, Howarth P et al. 2012. Mepolizumab for severe eosinophilic asthma (DREAM): a multicentre, double-blind, placebo-controlled trial. Lancet 380:9842651–59
    [Google Scholar]
  44. 44. 
    Chupp GL, Bradford ES, Albers FC et al. 2017. Efficacy of mepolizumab add-on therapy on health-related quality of life and markers of asthma control in severe eosinophilic asthma (MUSCA): a randomised, double-blind, placebo-controlled, parallel-group, multicentre, phase 3b trial. Lancet Respir. Med. 5:5390–400
    [Google Scholar]
  45. 45. 
    Wechsler ME, Akuthota P, Jayne D et al. 2017. Mepolizumab or placebo for eosinophilic granulomatosis with polyangiitis. N. Engl. J. Med. 376:201921–32
    [Google Scholar]
  46. 46. 
    Walsh GM. 2009. Reslizumab, a humanized anti-IL-5 mAb for the treatment of eosinophil-mediated inflammatory conditions. Curr. Opin. Mol. Ther. 11:3329–36
    [Google Scholar]
  47. 47. 
    Castro M, Zangrilli J, Wechsler ME et al. 2015. 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. 3:5355–66
    [Google Scholar]
  48. 48. 
    Weinstein SF, Katial RK, Bardin P et al. 2019. Effects of reslizumab on asthma outcomes in a subgroup of eosinophilic asthma patients with self-reported chronic rhinosinusitis with nasal polyps. J. Allergy Clin. Immunol. Practice 7:2589–96
    [Google Scholar]
  49. 49. 
    Brusselle G, Germinaro M, Weiss S, Zangrilli J 2017. Reslizumab in patients with inadequately controlled late-onset asthma and elevated blood eosinophils. Pulm. Pharmacol. Ther. 43:39–45
    [Google Scholar]
  50. 50. 
    Ghazi A, Trikha A, Calhoun WJ 2012. Benralizumab—a humanized mAb to IL-5Rα with enhanced antibody-dependent cell-mediated cytotoxicity—a novel approach for the treatment of asthma. Expert Opin. Biol. Ther. 12:1113–18
    [Google Scholar]
  51. 51. 
    Pham TH, Damera G, Newbold P, Ranade K 2016. Reductions in eosinophil biomarkers by benralizumab in patients with asthma. Respir. Med. 111:21–29
    [Google Scholar]
  52. 52. 
    Chupp G, Lugogo NL, Kline JN et al. 2019. Rapid onset of effect of benralizumab on morning peak expiratory flow in severe, uncontrolled asthma. Ann. Allergy Asthma Immunol. 122:5478–85
    [Google Scholar]
  53. 53. 
    Bleecker ER, FitzGerald JM, Chanez P et al. 2016. Efficacy and safety of benralizumab for patients with severe asthma uncontrolled with high-dosage inhaled corticosteroids and long-acting beta2-agonists (SIROCCO): a randomised, multicentre, placebo-controlled phase 3 trial. Lancet 388:100562115–27
    [Google Scholar]
  54. 54. 
    FitzGerald JM, Bleecker ER, Nair P et al. 2016. Benralizumab, an anti-interleukin-5 receptor alpha monoclonal antibody, as add-on treatment for patients with severe, uncontrolled, eosinophilic asthma (CALIMA): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet 388:100562128–41
    [Google Scholar]
  55. 55. 
    Nair P, Wenzel S, Rabe KF et al. 2017. Oral glucocorticoid-sparing effect of benralizumab in severe asthma. N. Engl. J. Med. 376:252448–58
    [Google Scholar]
  56. 56. 
    Santini G, Mores N, Malerba M et al. 2017. Dupilumab for the treatment of asthma. Expert Opin. Investig. Drugs 26:3357–66
    [Google Scholar]
  57. 57. 
    Rodrigues MA, Nogueira M, Torres T 2019. Dupilumab for atopic dermatitis. Evidence to date. G. Ital. Dermatol. Venereol. In press. https://doi.org/10.23736/S0392-0488.19.06417-4
    [Crossref] [Google Scholar]
  58. 58. 
    Bachert C, Hellings PW, Mullol J et al. 2019. Dupilumab improves patient-reported outcomes in patients with chronic rhinosinusitis with nasal polyps and comorbid asthma. J. Allergy Clin. Immunol. Practice 7:72447–49
    [Google Scholar]
  59. 59. 
    Weinstein SF, Katial R, Jayawardena S et al. 2018. Efficacy and safety of dupilumab in perennial allergic rhinitis and comorbid asthma. J. Allergy Clin. Immunol. 142:1171–77
    [Google Scholar]
  60. 60. 
    Simpson EL, Parnes JR, She D et al. 2019. Tezepelumab, an anti-thymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: a randomized phase 2a clinical trial. J. Am. Acad. Dermatol. 80:41013–21
    [Google Scholar]
  61. 61. 
    Erpenbeck VJ, Popov TA, Miller D et al. 2016. The oral CRTh2 antagonist QAW039 (fevipiprant): a phase II study in uncontrolled allergic asthma. Pulm. Pharmacol. Ther. 39:54–63
    [Google Scholar]
  62. 62. 
    Panettieri RA, Kotlikoff MI, Gerthoffer WT et al. 2008. Airway smooth muscle in bronchial tone, inflammation, and remodeling: basic knowledge to clinical relevance. Am. J. Respir. Crit. Care Med. 177:3248–52
    [Google Scholar]
  63. 63. 
    Prakash YS. 2016. Emerging concepts in smooth muscle contributions to airway structure and function: implications for health and disease. Am. J. Physiol. Lung Cell Mol. Physiol. 311:6L1113–L40
    [Google Scholar]
  64. 64. 
    Huber HL, Koessler KK. 1922. The pathology of bronchial asthma. Arch. Intern. Med. 30:6689–760
    [Google Scholar]
  65. 65. 
    Bergeron C, Boulet LP. 2006. Structural changes in airway diseases: characteristics, mechanism, consequences, and pharmacologic modulation. Chest 129:1068–87
    [Google Scholar]
  66. 66. 
    Kuwano K, Bosken CH, Paré PD et al. 1993. Small airways dimensions in asthma and in chronic obstructive pulmonary disease. Am. Rev. Respir. Dis. 148:51220–25
    [Google Scholar]
  67. 67. 
    Roche WR, Beasley R, Williams JH et al. 1989. Subepithelial fibrosis in the bronchi of asthmatics. Lancet 1:8637520–24
    [Google Scholar]
  68. 68. 
    Brewster CE, Howarth PH, Djukanovic R et al. 1990. Myofibroblasts and subepithelial fibrosis in bronchial asthma. Am. J. Respir. Cell Mol. Biol. 3:5507–11
    [Google Scholar]
  69. 69. 
    Benayoun L, Druilhe A, Dombret MC et al. 2003. Airway structural alterations selectively associated with severe asthma. Am. J. Respir. Crit. Care Med. 167:101360–68
    [Google Scholar]
  70. 70. 
    Elias JA, Zhu Z, Chupp G, Homer RJ 1999. Airway remodeling in asthma. J. Clin. Investig. 104:81001–6
    [Google Scholar]
  71. 71. 
    Ni Y, Mulier S, Maio Y et al. 2005. A review of the general aspects of radiofrequency ablation. Abdom. Imaging 30:4381–400
    [Google Scholar]
  72. 72. 
    Danek CJ, Lombard CM, Dungworth DL et al. 2004. Reduction in airway hyperresponsiveness to methacholine by the application of RF energy in dogs. J. Appl. Physiol. 97:51946–53
    [Google Scholar]
  73. 73. 
    Miller JD, Cox G, Vincic L et al. 2005. A prospective feasibility study of bronchial thermoplasty in the human airway. Chest 127:61999–2006
    [Google Scholar]
  74. 74. 
    Cox G, Miller JD, McWilliams A et al. 2006. Bronchial thermoplasty for asthma. Am. J. Respir. Crit. Care Med. 173:9965–69
    [Google Scholar]
  75. 75. 
    Castro M, Rubin AS, Laviolette M et al. 2010. Effectiveness and safety of bronchial thermoplasty in the treatment of severe asthma: a multicenter, randomized, double-blind, sham-controlled clinical trial. Am. J. Respir. Crit. Care Med. 181:2116–24
    [Google Scholar]
  76. 76. 
    Chupp G, Laviolette M, Cohn L et al. 2017. Long-term outcomes of bronchial thermoplasty in subjects with severe asthma: a comparison of 3-year follow-up results from two prospective multicentre studies. Eur. Respir. J. 50:21750017
    [Google Scholar]
  77. 77. 
    Pretolani M, Berggvist A, Thabut G et al. 2017. Effectiveness of bronchial thermoplasty in patients with severe refractory asthma: clinical and histopathologic correlations. J. Allergy Clin. Immunol. 139:41176–85
    [Google Scholar]
  78. 78. 
    Salem IH, Boulet LP, Biardel S et al. 2016. Long-term effects of bronchial thermoplasty on airway smooth muscle and reticular basement membrane thickness in severe asthma. Ann. Am. Thorac. Soc. 13:81426–28
    [Google Scholar]
  79. 79. 
    Facciolongo N, Di Stefano A, Pietrini V et al. 2018. Nerve ablation after bronchial thermoplasty and sustained improvement in severe asthma. BMC Pulm. Med. 18:129
    [Google Scholar]
  80. 80. 
    Blaiss MS, Castro M, Chipps BE et al. 2017. Guiding principles for use of newer biologic and bronchial thermoplasty for patients with severe asthma. Ann. Allergy Asthma Immunol. 119:6533–40
    [Google Scholar]
  81. 81. 
    Trivedi A, Pavord ID, Castro M 2016. Bronchial thermoplasty and biological therapy as targeted treatments for severe uncontrolled asthma. Lancet 4:7585–92
    [Google Scholar]
  82. 82. 
    Zein JG, Menegay MC, Singer ME et al. 2016. Cost effectiveness of bronchial thermoplasty in patients with severe uncontrolled asthma. J. Asthma 53:2194–200
    [Google Scholar]
  83. 83. 
    Cangelosi MJ, Ortendahl JD, Meckley LM et al. 2015. Cost-effectiveness of bronchial thermoplasty in commercially-insured patients with poorly controlled, severe, persistent asthma. Expert Rev. Pharmacoecon. Outcomes Res. 15:2357–64
    [Google Scholar]
  84. 84. 
    Zafari ZSM, Marra C, Chen W, FitzGerald JM 2016. Cost-effectiveness of bronchial thermoplasty, omalizumab, and standard therapy for moderate-to-severe allergic asthma. PLOS ONE 11:1e0146003
    [Google Scholar]
/content/journals/10.1146/annurev-med-041818-020630
Loading
/content/journals/10.1146/annurev-med-041818-020630
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error