Allergy Asthma Respir Dis.  2016 Jan;4(1):4-13. 10.4168/aard.2016.4.1.4.

Biomarkers of adult asthma and personalized medicine

Affiliations
  • 1Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea. jhleemd@yuhs.ac

Abstract

The concept of personalized medicine for disease diagnosis, treatment, and management, considering individual variability, including susceptibility, clinical manifestations, and drug responsiveness, is a global emerging trend in medicine, which is also inevitable. However, clinical applications of personalized medicine in the real-world practice have been limited to certain cancers so far. Furthermore, this new concept to the diagnosis and treatment of adult asthma has not been applied to clinical use. Asthma is a multifactorial and heterogeneous disease. It seems to encompass a broad spectrum of clinical manifestations with different underlying pathophysiological mechanisms. Thus, it is not easy to categorize by their clinical features alone. Endotypical categorization that considering specific pathophysiological mechanisms will be more helpful in applying the concept of personalized medicine. The success of personalized medicine depends on patient selection for precise prescription of asthma medications. In the recent years, many investigators and physicians have devoted a lot of effort to the discovery of reliable biomarkers in asthmatic patients, which will be able to actualize the personalized medicine in near future. Despite such great efforts toward investigation of good biomarkers, few things have turned out to be practical in the clinic. Easily interpretable biomarkers of asthma are necessary to assess early detection, determination of treatment, prognosis prediction, and monitoring of exacerbation. Herein, we review recent studies regarding disease classifications and biomarkers of asthma.

Keyword

Asthma; Biomarkers; Phenotype; Endotype; Personalized medicine

MeSH Terms

Adult*
Asthma*
Biomarkers*
Classification
Diagnosis
Humans
Precision Medicine*
Patient Selection
Phenotype
Prescriptions
Prognosis
Research Personnel

Figure

  • Fig. 1 The schematic presentation of the underlying pathogenesis of allergic asthma. (A) The progress of allergic asthma demanded sensitization to an environmental allergen. Primary exposure to an allergen conducted activation of Th2 lymphocytes and ILC2. (B) Subsequently, re-exposure causes immediate reaction of the increasing production of Th2 cytokines, stimulation of IgE synthesis and accumulation of inflammatory cells in airway. PRR, pattern recognition receptor; IL, interleukin; ILC2, type 2 innate lymphoid cell; DC, dendritic cell.


Cited by  2 articles

Current status of microbiome research in asthma and chronic obstructive pulmonary disease
Byung-Keun Kim, Chin Kook Rhee, Ji Ye Jung, Hye-Ryun Kang, Sang-Heon Cho
Allergy Asthma Respir Dis. 2016;4(5):321-327.    doi: 10.4168/aard.2016.4.5.321.

Could exhaled breath temperature discriminate the asthma?
Man Yong Han
Allergy Asthma Respir Dis. 2017;5(3):121-122.    doi: 10.4168/aard.2017.5.3.121.


Reference

1. Lai CK, Beasley R, Crane J, Foliaki S, Shah J, Weiland S, et al. Global variation in the prevalence and severity of asthma symptoms: phase three of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax. 2009; 64:476–483.
Article
2. Kim CY, Park HW, Ko SK, Chang SI, Moon HB, Kim YY, et al. The financial burden of asthma: a nationwide comprehensive survey conducted in the republic of Korea. Allergy Asthma Immunol Res. 2011; 3:34–38.
Article
3. Anandan C, Nurmatov U, van Schayck OC, Sheikh A. Is the prevalence of asthma declining? Systematic review of epidemiological studies. Allergy. 2010; 65:152–167.
Article
4. Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med. 2010; 181:315–323.
Article
5. Sutherland ER, Goleva E, King TS, Lehman E, Stevens AD, Jackson LP, et al. Cluster analysis of obesity and asthma phenotypes. PLoS One. 2012; 7:e36631.
Article
6. Hamburg MA, Collins FS. The path to personalized medicine. N Engl J Med. 2010; 363:301–304.
Article
7. Lipworth BJ. Systemic adverse effects of inhaled corticosteroid therapy: a systematic review and meta-analysis. Arch Intern Med. 1999; 159:941–955.
Article
8. Wenzel S. Severe asthma in adults. Am J Respir Crit Care Med. 2005; 172:149–160.
Article
9. Busse WW. Asthma diagnosis and treatment: filling in the information gaps. J Allergy Clin Immunol. 2011; 128:740–750.
Article
10. Soukhanov AH. Encarta world English dictionary. New York: St Martin's Press;1999.
11. Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012; 18:716–725.
Article
12. Lee HY, Kang JY, Yoon HK, Lee SY, Kwon SS, Kim YK, et al. Clinical characteristics of asthma combined with COPD feature. Yonsei Med J. 2014; 55:980–986.
Article
13. Kim TB, Jang AS, Kwon HS, Park JS, Chang YS, Cho SH, et al. Identification of asthma clusters in two independent Korean adult asthma cohorts. Eur Respir J. 2013; 41:1308–1314.
Article
14. Bourdin A, Molinari N, Vachier I, Varrin M, Marin G, Gamez AS, et al. Prognostic value of cluster analysis of severe asthma phenotypes. J Allergy Clin Immunol. 2014; 134:1043–1050.
Article
15. Fingleton J, Travers J, Williams M, Charles T, Bowles D, Strik R, et al. Treatment responsiveness of phenotypes of symptomatic airways obstruction in adults. J Allergy Clin Immunol. 2015; 136:601–609.
Article
16. Corren J. Asthma phenotypes and endotypes: an evolving paradigm for classification. Discov Med. 2013; 15:243–249.
17. Bartminski G, Crossley M, Turcanu V. Novel biomarkers for asthma stratification and personalized therapy. Expert Rev Mol Diagn. 2015; 15:415–430.
Article
18. Anderson GP. Endotyping asthma: new insights into key pathogenic mechanisms in a complex, heterogeneous disease. Lancet. 2008; 372:1107–1119.
Article
19. De Gruttola VG, Clax P, DeMets DL, Downing GJ, Ellenberg SS, Friedman L, et al. Considerations in the evaluation of surrogate endpoints in clinical trials. summary of a National Institutes of Health workshop. Control Clin Trials. 2001; 22:485–502.
Article
20. Bousquet J, Chanez P, Lacoste JY, Barnéon G, Ghavanian N, Enander I, et al. Eosinophilic inflammation in asthma. N Engl J Med. 1990; 323:1033–1039.
Article
21. Marko-Varga GA, Nilsson J, Laurell T. New directions of miniaturization within the biomarker research area. Electrophoresis. 2004; 25:3479–3491.
Article
22. Deckers J, Branco Madeira F, Hammad H. Innate immune cells in asthma. Trends Immunol. 2013; 34:540–547.
Article
23. Cayrol C, Girard JP. IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. Curr Opin Immunol. 2014; 31:31–37.
Article
24. Suzukawa M, Morita H, Nambu A, Arae K, Shimura E, Shibui A, et al. Epithelial cell-derived IL-25, but not Th17 cell-derived IL-17 or IL-17F, is crucial for murine asthma. J Immunol. 2012; 189:3641–3652.
Article
25. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+) Sca-1(+) lymphoid cells. Nature. 2010; 463:540–544.
Article
26. Sibilano R, Frossi B, Pucillo CE. Mast cell activation: a complex interplay of positive and negative signaling pathways. Eur J Immunol. 2014; 44:2558–2566.
Article
27. Bochner BS, Busse WW. Allergy and asthma. J Allergy Clin Immunol. 2005; 115:953–959.
Article
28. Jacobsen EA, Zellner KR, Colbert D, Lee NA, Lee JJ. Eosinophils regulate dendritic cells and Th2 pulmonary immune responses following allergen provocation. J Immunol. 2011; 187:6059–6068.
Article
29. Schuijs MJ, Willart MA, Hammad H, Lambrecht BN. Cytokine targets in airway inflammation. Curr Opin Pharmacol. 2013; 13:351–361.
Article
30. Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009; 180:388–395.
Article
31. Nakagome K, Matsushita S, Nagata M. Neutrophilic inflammation in severe asthma. Int Arch Allergy Immunol. 2012; 158:Suppl 1. 96–102.
Article
32. Durrant DM, Metzger DW. Emerging roles of T helper subsets in the pathogenesis of asthma. Immunol Invest. 2010; 39:526–549.
Article
33. Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A, et al. Mepolizumab and exacerbations of refractory eosinophilic asthma. N Engl J Med. 2009; 360:973–984.
Article
34. Lim S, Jatakanon A, Meah S, Oates T, Chung KF, Barnes PJ. Relationship between exhaled nitric oxide and mucosal eosinophilic inflammation in mild to moderately severe asthma. Thorax. 2000; 55:184–188.
Article
35. Crimi E, Spanevello A, Neri M, Ind PW, Rossi GA, Brusasco V. Dissociation between airway inflammation and airway hyperresponsiveness in allergic asthma. Am J Respir Crit Care Med. 1998; 157:4–9.
Article
36. Loutsios C, Farahi N, Porter L, Lok LS, Peters AM, Condliffe AM, et al. Biomarkers of eosinophilic inflammation in asthma. Expert Rev Respir Med. 2014; 8:143–150.
Article
37. Halonen M, Barbee RA, Lebowitz MD, Burrows B. An epidemiologic study of interrelationships of total serum immunoglobulin E, allergy skin-test reactivity, and eosinophilia. J Allergy Clin Immunol. 1982; 69:221–228.
Article
38. Sherrill DL, Lebowitz MD, Halonen M, Barbee RA, Burrows B. Longitudinal evaluation of the association between pulmonary function and total serum IgE. Am J Respir Crit Care Med. 1995; 152:98–102.
Article
39. Simpson A, Soderstrom L, Ahlstedt S, Murray CS, Woodcock A, Custovic A. IgE antibody quantification and the probability of wheeze in preschool children. J Allergy Clin Immunol. 2005; 116:744–749.
Article
40. Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013; 368:2455–2466.
Article
41. Corren J, Lemanske RF, Hanania NA, Korenblat PE, Parsey MV, Arron JR, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med. 2011; 365:1088–1098.
Article
42. Szefler SJ, Wenzel S, Brown R, Erzurum SC, Fahy JV, Hamilton RG, et al. Asthma outcomes: biomarkers. J Allergy Clin Immunol. 2012; 129:3 Suppl. S9–23.
Article
43. Baldacci S, Omenaas E, Oryszczyn MP. Allergy markers in respiratory epidemiology. Eur Respir J. 2001; 17:773–790.
Article
44. Hastie AT, Moore WC, Li H, Rector BM, Ortega VE, Pascual RM, et al. Biomarker surrogates do not accurately predict sputum eosinophil and neutrophil percentages in asthmatic subjects. J Allergy Clin Immunol. 2013; 132:72–80.
Article
45. Druilhe A, Létuvé S, Pretolani M. Glucocorticoid-induced apoptosis in human eosinophils: mechanisms of action. Apoptosis. 2003; 8:481–495.
46. Massanari M, Holgate ST, Busse WW, Jimenez P, Kianifard F, Zeldin R. Effect of omalizumab on peripheral blood eosinophilia in allergic asthma. Respir Med. 2010; 104:188–196.
Article
47. Castro M, Mathur S, Hargreave F, Boulet LP, Xie F, Young J, et al. Reslizumab for poorly controlled, eosinophilic asthma: a randomized, placebo-controlled study. Am J Respir Crit Care Med. 2011; 184:1125–1132.
Article
48. Leckie MJ, ten Brinke A, Khan J, Diamant Z, O'Connor BJ, Walls CM, et al. Effects of an interleukin-5 blocking monoclonal antibody on eosinophils, airway hyper-responsiveness, and the late asthmatic response. Lancet. 2000; 356:2144–2148.
Article
49. Valent P, Gleich GJ, Reiter A, Roufosse F, Weller PF, Hellmann A, et al. Pathogenesis and classification of eosinophil disorders: a review of recent developments in the field. Expert Rev Hematol. 2012; 5:157–176.
Article
50. Connett GJ. Bronchoalveolar lavage. Paediatr Respir Rev. 2000; 1:52–56.
Article
51. 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.
52. Kips JC, Fahy JV, Hargreave FE, Ind PW, in't Veen JC. Methods for sputum induction and analysis of induced sputum: a method for assessing airway inflammation in asthma. Eur Respir J Suppl. 1998; 26:9S–12S.
53. Simpson JL, Scott R, Boyle MJ, Gibson PG. Inflammatory subtypes in asthma: assessment and identification using induced sputum. Respirology. 2006; 11:54–61.
Article
54. Green RH, Brightling CE, McKenna S, Hargadon B, Parker D, Bradding P, et al. Asthma exacerbations and sputum eosinophil counts: a randomised controlled trial. Lancet. 2002; 360:1715–1721.
Article
55. Berry M, Morgan A, Shaw DE, Parker D, Green R, Brightling C, et al. Pathological features and inhaled corticosteroid response of eosinophilic and non-eosinophilic asthma. Thorax. 2007; 62:1043–1049.
Article
56. Fleming L, Wilson N, Regamey N, Bush A. Use of sputum eosinophil counts to guide management in children with severe asthma. Thorax. 2012; 67:193–198.
Article
57. Woodruff PG, Khashayar R, Lazarus SC, Janson S, Avila P, Boushey HA, et al. Relationship between airway inflammation, hyperresponsiveness, and obstruction in asthma. J Allergy Clin Immunol. 2001; 108:753–758.
Article
58. Lex C, Ferreira F, Zacharasiewicz A, Nicholson AG, Haslam PL, Wilson NM, et al. Airway eosinophilia in children with severe asthma: predictive values of noninvasive tests. Am J Respir Crit Care Med. 2006; 174:1286–1291.
59. Morris SM Jr, Billiar TR. New insights into the regulation of inducible nitric oxide synthesis. Am J Physiol. 1994; 266(6 Pt 1):E829–E839.
Article
60. Dweik RA, Boggs PB, Erzurum SC, Irvin CG, Leigh MW, Lundberg JO, 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; 184:602–615.
Article
61. Amelink M, de Groot JC, de Nijs SB, Lutter R, Zwinderman AH, Sterk PJ, et al. Severe adult-onset asthma: a distinct phenotype. J Allergy Clin Immunol. 2013; 132:336–341.
Article
62. 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; 186:1102–1108.
Article
63. Smith AD, Cowan JO, Brassett KP, Herbison GP, Taylor DR. Use of exhaled nitric oxide measurements to guide treatment in chronic asthma. N Engl J Med. 2005; 352:2163–2173.
Article
64. Petsky HL, Cates CJ, Lasserson TJ, Li AM, Turner C, Kynaston JA, et al. A systematic review and meta-analysis: tailoring asthma treatment on eosinophilic markers (exhaled nitric oxide or sputum eosinophils). Thorax. 2012; 67:199–208.
Article
65. Calhoun WJ, Ameredes BT, King TS, Icitovic N, Bleecker ER, Castro M, et al. Comparison of physician-, biomarker-, and symptom-based strategies for adjustment of inhaled corticosteroid therapy in adults with asthma: the BASALT randomized controlled trial. JAMA. 2012; 308:987–997.
Article
66. Deykin A. Targeting biologic markers in asthma--is exhaled nitric oxide the bull's-eye? N Engl J Med. 2005; 352:2233–2235.
Article
67. Gillan L, Matei D, Fishman DA, Gerbin CS, Karlan BY, Chang DD. Periostin secreted by epithelial ovarian carcinoma is a ligand for alpha(V)beta(3) and alpha(V)beta(5) integrins and promotes cell motility. Cancer Res. 2002; 62:5358–5364.
68. Sidhu SS, Yuan S, Innes AL, Kerr S, Woodruff PG, Hou L, et al. Roles of epithelial cell-derived periostin in TGF-beta activation, collagen production, and collagen gel elasticity in asthma. Proc Natl Acad Sci U S A. 2010; 107:14170–14175.
Article
69. Woodruff PG, Boushey HA, Dolganov GM, Barker CS, Yang YH, Donnelly S, et al. Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci U S A. 2007; 104:15858–15863.
Article
70. Hanania NA, Wenzel S, Rosén K, Hsieh HJ, Mosesova S, Choy DF, 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; 187:804–811.
Article
71. Cai C, Yang J, Hu S, Zhou M, Guo W. Relationship between urinary cysteinyl leukotriene E4 levels and clinical response to antileukotriene treatment in patients with asthma. Lung. 2007; 185:105–112.
Article
72. Chupp GL, Lee CG, Jarjour N, Shim YM, Holm CT, He S, et al. A chitinase-like protein in the lung and circulation of patients with severe asthma. N Engl J Med. 2007; 357:2016–2027.
Article
73. Konradsen JR, James A, Nordlund B, Reinius LE, Söderhäll C, Melön E, et al. The chitinase-like protein YKL-40: a possible biomarker of inflammation and airway remodeling in severe pediatric asthma. J Allergy Clin Immunol. 2013; 132:328–335.e5.
74. Specjalski K, Chełmińska M, Jassem E. YKL-40 protein correlates with the phenotype of asthma. Lung. 2015; 193:189–194.
Article
75. Ober C, Tan Z, Sun Y, Possick JD, Pan L, Nicolae R, et al. Effect of variation in CHI3L1 on serum YKL-40 level, risk of asthma, and lung function. N Engl J Med. 2008; 358:1682–1691.
Article
76. Fens N, Roldaan AC, van der Schee MP, Boksem RJ, Zwinderman AH, Bel EH, et al. External validation of exhaled breath profiling using an electronic nose in the discrimination of asthma with fixed airways obstruction and chronic obstructive pulmonary disease. Clin Exp Allergy. 2011; 41:1371–1378.
Article
77. Dragonieri S, Schot R, Mertens BJ, Le Cessie S, Gauw SA, Spanevello A, et al. An electronic nose in the discrimination of patients with asthma and controls. J Allergy Clin Immunol. 2007; 120:856–862.
Article
78. van der Schee MP, Palmay R, Cowan JO, Taylor DR. Predicting steroid responsiveness in patients with asthma using exhaled breath profiling. Clin Exp Allergy. 2013; 43:1217–1225.
Article
79. Horvath I, Hunt J, Barnes PJ, Alving K, Antczak A, Baraldi E, et al. Exhaled breath condensate: methodological recommendations and unresolved questions. Eur Respir J. 2005; 26:523–548.
Article
80. Tufvesson E, Bjermer L. Methodological improvements for measuring eicosanoids and cytokines in exhaled breath condensate. Respir Med. 2006; 100:34–38.
Article
81. Montuschi P, Barnes PJ. Exhaled leukotrienes and prostaglandins in asthma. J Allergy Clin Immunol. 2002; 109:615–620.
Article
82. Fireman E, Shtark M, Priel IE, Shiner R, Mor R, Kivity S, et al. Hydrogen peroxide in exhaled breath condensate (EBC) vs eosinophil count in induced sputum (IS) in parenchymal vs airways lung diseases. Inflammation. 2007; 30:44–51.
Article
83. Koh GC, Shek LP, Goh DY, Van Bever H, Koh DS. Eosinophil cationic protein: is it useful in asthma? A systematic review. Respir Med. 2007; 101:696–705.
Article
84. Bartoli ML, Bacci E, Carnevali S, Cianchetti S, Dente FL, Di Franco A, et al. Clinical assessment of asthma severity partially corresponds to sputum eosinophilic airway inflammation. Respir Med. 2004; 98:184–193.
Article
85. Prehn A, Seger RA, Torresani T, Molinari L, Sennhauser FH. Evaluation of a clinical algorithm involving serum eosinophil cationic protein for guiding the anti-inflammatory treatment of bronchial asthma in childhood. Pediatr Allergy Immunol. 2000; 11:87–94.
Article
86. Kumar SD, Emery MJ, Atkins ND, Danta I, Wanner A. Airway mucosal blood flow in bronchial asthma. Am J Respir Crit Care Med. 1998; 158:153–156.
Article
87. García G, Bergna M, Uribe E, Yañez A, Soriano JB. Increased exhaled breath temperature in subjects with uncontrolled asthma. Int J Tuberc Lung Dis. 2013; 17:969–972.
Article
88. Chan IS, Ginsburg GS. Personalized medicine: progress and promise. Annu Rev Genomics Hum Genet. 2011; 12:217–244.
Article
89. Cowan DC, Taylor DR, Peterson LE, Cowan JO, Palmay R, Williamson A, et al. Biomarker-based asthma phenotypes of corticosteroid response. J Allergy Clin Immunol. 2015; 135:877–883.e1.
Article
Full Text Links
  • AARD
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr