The value of blood cytokines and chemokines in assessing COPD

Eric Bradford, Sean Jacobson, Jason Varasteh, Alejandro P Comellas, Prescott Woodruff, Wanda O'Neal, Dawn L DeMeo, Xingnan Li, Victor Kim, Michael Cho, Peter J Castaldi, Craig Hersh, Edwin K Silverman, James D Crapo, Katerina Kechris, Russell P Bowler, Eric Bradford, Sean Jacobson, Jason Varasteh, Alejandro P Comellas, Prescott Woodruff, Wanda O'Neal, Dawn L DeMeo, Xingnan Li, Victor Kim, Michael Cho, Peter J Castaldi, Craig Hersh, Edwin K Silverman, James D Crapo, Katerina Kechris, Russell P Bowler

Abstract

Background: Blood biomarkers are increasingly used to stratify high risk chronic obstructive pulmonary disease (COPD) patients; however, there are fewer studies that have investigated multiple biomarkers and replicated in multiple large well-characterized cohorts of susceptible current and former smokers.

Methods: We used two MSD multiplex panels to measure 9 cytokines and chemokines in 2123 subjects from COPDGene and 1117 subjects from SPIROMICS. These biomarkers included: interleukin (IL)-2, IL-6, IL-8, IL-10, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, eotaxin/CCL-11, eotaxin-3/CCL-26, and thymus and activation-regulated chemokine (TARC)/CCL-17. Regression models adjusted for clinical covariates were used to determine which biomarkers were associated with the following COPD phenotypes: airflow obstruction (forced expiratory flow at 1 s (FEV1%) and FEV1/forced vital capacity (FEV1/FVC), chronic bronchitis, COPD exacerbations, and emphysema. Biomarker-genotype associations were assessed by genome-wide association of single nucleotide polymorphisms (SNPs).

Results: Eotaxin and IL-6 were strongly associated with airflow obstruction and accounted for 3-5% of the measurement variance on top of clinical variables. IL-6 was associated with progressive airflow obstruction over 5 years and both IL-6 and IL-8 were associated with progressive emphysema over 5 years. None of the biomarkers were consistently associated with chronic bronchitis or COPD exacerbations. We identified one novel SNP (rs9302690 SNP) that was associated with CCL17 plasma measurements.

Conclusion: When assessing smoking related pulmonary disease, biomarkers of inflammation such as IL-2, IL-6, IL-8, and eotaxin may add additional modest predictive value on top of clinical variables alone.

Trial registration: COPDGene (ClinicalTrials.gov Identifier: NCT02445183 ). Subpopulations and Intermediate Outcomes Measures in COPD Study (SPIROMICS) ( ClinicalTrials.gov Identifier: NCT 01969344 ).

Trial registration: ClinicalTrials.gov NCT02445183 NCT01969344.

Conflict of interest statement

Ethics approval and consent to participate

The study protocols were approved by the local ethics committees listed below. Informed written consent was obtained from all participants.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Plasma eotaxin and IL-6 are higher in subjects with COPD. Never smokers (never) and current and former smokers with no COPD (control), mild/moderate COPD (Mild/Moderate), or severe or very severe COPD (Severe)
Fig. 2
Fig. 2
Heat map showing associations between cytokines and chemokines and COPD clinical phenotypes in the COPDGene and SPIROMICS cohorts. The intensity of the color represents the log of the P-value with red indicating positive associations and blue indicating negative associations

References

    1. United States. Public Health Service. Office Of The Surgeon General . The health consequences of smoking--50 years of progress : a report of the surgeon general. Rockville: U.S. Department Of Health And Human Services, Public Health Service, Office Of The Surgeon General; 2014.
    1. Vestbo J, Agusti A, Wouters E, et al. Should we view chronic obstructive pulmonary disease differently after Eclipse? A clinical perspective from the study team. Am J Respir Crit Care Med. 2014;189(9):1022–1030. doi: 10.1164/rccm.201311-2006PP.
    1. Agusti A, Sin DD. Biomarkers in COPD. Clin Chest Med. 2014;35(1):131–141. doi: 10.1016/j.ccm.2013.09.006.
    1. Faner R, Tal-Singer R, Riley JH, et al. Lessons from Eclipse: a review of COPD biomarkers. Thorax. 2014;69(7):666–672. doi: 10.1136/thoraxjnl-2013-204778.
    1. Agusti A, Gea J, Faner R. Biomarkers, the control panel and personalized COPD medicine. Respirology. 2016;21(1):24–33. doi: 10.1111/resp.12585.
    1. Dahl M, Tybjaerg-Hansen A, Vestbo J, Lange P, Nordestgaard BG. Elevated plasma fibrinogen associated with reduced pulmonary function and increased risk of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164(6):1008–1011. doi: 10.1164/ajrccm.164.6.2010067.
    1. Dahl M, Vestbo J, Lange P, Bojesen SE, Tybjaerg-Hansen A, Nordestgaard BG. C-reactive protein as a predictor of prognosis in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007;175(3):250–255. doi: 10.1164/rccm.200605-713OC.
    1. De Torres JP, Cordoba-Lanus E, Lopez-Aguilar C, et al. C-reactive protein levels and clinically important predictive outcomes in stable COPD patients. Eur Respir J. 2006;27(5):902–907. doi: 10.1183/09031936.06.00109605.
    1. Agusti A, Edwards LD, Rennard SI, et al. Persistent systemic inflammation is associated with poor clinical outcomes in COPD: a novel phenotype. PLoS One. 2012;7(5):E37483. doi: 10.1371/journal.pone.0037483.
    1. Celli BR, Locantore N, Yates J, et al. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185(10):1065–1072. doi: 10.1164/rccm.201110-1792OC.
    1. Papaioannou AI, Mazioti A, Kiropoulos T, et al. Systemic and airway inflammation and the presence of emphysema in patients with COPD. Respir Med. 2010;104(2):275–282. doi: 10.1016/j.rmed.2009.09.016.
    1. Duvoix A, Dickens J, Haq I, et al. Blood fibrinogen as a biomarker of chronic obstructive pulmonary disease. Thorax. 2013;68(7):670–676. doi: 10.1136/thoraxjnl-2012-201871.
    1. Mannino DM, Valvi D, Mullerova H, Tal-Singer R. Fibrinogen, COPD and mortality in a nationally representative U.S. cohort. COPD. 2012;9(4):359–366. doi: 10.3109/15412555.2012.668249.
    1. Vestbo J, Edwards LD, Scanlon PD, et al. Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med. 2011;365(13):1184–1192. doi: 10.1056/NEJMoa1105482.
    1. Wedzicha JA, Seemungal TA, Maccallum PK, et al. Acute exacerbations of chronic obstructive pulmonary disease are accompanied by elevations of plasma fibrinogen and serum Il-6 levels. Thromb Haemost. 2000;84(2):210–215.
    1. Hurst JR, Vestbo J, Anzueto A, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363(12):1128–1138. doi: 10.1056/NEJMoa0909883.
    1. Thomsen M, Ingebrigtsen TS, Marott JL, et al. Inflammatory biomarkers and exacerbations in chronic obstructive pulmonary disease. JAMA. 2013;309(22):2353–2361. doi: 10.1001/jama.2013.5732.
    1. Eagan TM, Ueland T, Wagner PD, et al. Systemic inflammatory markers in COPD: results from the Bergen COPD cohort study. Eur Respir J. 2010;35(3):540–548. doi: 10.1183/09031936.00088209.
    1. Fogarty AW, Jones S, Britton JR, Lewis SA, Mckeever TM. Systemic inflammation and decline in lung function in a general population: a prospective study. Thorax. 2007;62(6):515–520. doi: 10.1136/thx.2006.066969.
    1. Engstrom G, Segelstorm N, Ekberg-Aronsson M, Nilsson PM, Lindgarde F, Lofdahl CG. Plasma markers of inflammation and incidence of Hospitalisations for COPD: results from a population-based cohort study. Thorax. 2009;64(3):211–215. doi: 10.1136/thx.2008.102079.
    1. Mannino DM, Tal-Singer R, Lomas DA, et al. Plasma fibrinogen as a biomarker for mortality and hospitalized exacerbations in people with COPD. Chronic Obstr Pulm Dis (Miami) 2015;2(1):23–34. doi: 10.15326/jcopdf.2.1.2014.0138.
    1. Cockayne DA, Cheng DT, Waschki B, et al. Systemic biomarkers of neutrophilic inflammation, tissue injury and repair in COPD patients with differing levels of disease severity. PLoS One. 2012;7(6):E38629. doi: 10.1371/journal.pone.0038629.
    1. Cheng DT, Kim DK, Cockayne DA, et al. Systemic soluble receptor for advanced Glycation Endproducts is a biomarker of emphysema and associated with ager genetic variants in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;188(8):948–957. doi: 10.1164/rccm.201302-0247OC.
    1. Carolan BJ, Hughes G, Morrow J, et al. The association of plasma biomarkers with computed tomography-assessed emphysema phenotypes. Respir Res. 2014;15:127. doi: 10.1186/s12931-014-0127-9.
    1. Coxson HO, Dirksen A, Edwards LD, et al. The presence and progression of emphysema in COPD as determined by ct scanning and biomarker expression: a prospective analysis from the Eclipse study. Lancet Respir Med. 2013;1(2):129–136. doi: 10.1016/S2213-2600(13)70006-7.
    1. Smith DJ, Yerkovich ST, Towers MA, Carroll ML, Thomas R, Upham JW. Reduced soluble receptor for advanced glycation end-products in COPD. Eur Respir J. 2011;37(3):516–522. doi: 10.1183/09031936.00029310.
    1. Kobayashi H, Kanoh S, Motoyoshi K. Serum surfactant protein-A, but not surfactant protein-D or KL-6, can predict preclinical lung damage induced by smoking. Biomarkers. 2008;13(4):385–392. doi: 10.1080/13547500801903651.
    1. Winkler C, Atochina-Vasserman EN, Holz O, et al. Comprehensive characterisation of pulmonary and serum surfactant protein D in COPD. Respir Res. 2011;12:29. doi: 10.1186/1465-9921-12-29.
    1. Lomas DA, Silverman EK, Edwards LD, et al. Serum surfactant protein D is steroid sensitive and associated with exacerbations of COPD. Eur Respir J. 2009;34(1):95–102. doi: 10.1183/09031936.00156508.
    1. Lomas DA, Silverman EK, Edwards LD, Miller BE, Coxson HO, Tal-Singer R. Evaluation of serum Cc-16 as a biomarker for COPD in the Eclipse cohort. Thorax. 2008;63(12):1058–1063. doi: 10.1136/thx.2008.102574.
    1. Regan EA, Hokanson JE, Murphy JR, et al. Genetic epidemiology of COPD (COPDGene) study design. COPD. 2010;7(1):32–43. doi: 10.3109/15412550903499522.
    1. Couper D, Lavange LM, Han M, et al. Design of the subpopulations and intermediate outcomes in COPD study (Spiromics) Thorax. 2014;69(5):491–494. doi: 10.1136/thoraxjnl-2013-203897.
    1. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS, Committee GS Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. Nhlbi/who global initiative for chronic obstructive lung disease (GOLD) workshop summary. Am J Respir Crit Care Med. 2001;163(5):1256–1276. doi: 10.1164/ajrccm.163.5.2101039.
    1. Sun W, Kechris K, Jacobson S, et al. Common genetic polymorphisms influence blood biomarker measurements in COPD. PLoS Genet. 2016;12(8):E1006011. doi: 10.1371/journal.pgen.1006011.
    1. Celli BR, Cote CG, Marin JM, et al. The body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med. 2004;350(10):1005–1012. doi: 10.1056/NEJMoa021322.
    1. Puhan MA, Garcia-Aymerich J, Frey M, et al. Expansion of the prognostic assessment of patients with chronic obstructive pulmonary disease: the updated Bode index and the ado index. Lancet. 2009;374(9691):704–711. doi: 10.1016/S0140-6736(09)61301-5.
    1. Mcfadden D. Conditional logit analysis of qualitative choice behavior. In: Zarembka P, Ed. Frontiers in econometrics. Cambridge: Elsevier; 1974:105-142.
    1. Cragg JG. The demand for automobiles. Can J Econ. 1970;3:386–406. doi: 10.2307/133656.
    1. Nakagawa S, Schielzeth H. A general and simple method for obtaining R2 from generalized linear mixed-effects models. Methods Ecol Evol. 2013;4(2):133–142. doi: 10.1111/j.2041-210x.2012.00261.x.
    1. Han MK, Agusti A, Calverley PM, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182(5):598–604. doi: 10.1164/rccm.200912-1843CC.
    1. Lilly CM, Woodruff PG, Camargo CA, Jr, et al. Elevated plasma eotaxin levels in patients with acute asthma. J Allergy Clin Immunol. 1999;104(4 Pt 1):786–790. doi: 10.1016/S0091-6749(99)70288-5.
    1. Adar T, Shteingart S, Ben Ya'acov A, Bar-Gil Shitrit A, Goldin E. From airway inflammation to inflammatory bowel disease: eotaxin-1, a key regulator of intestinal inflammation. Clin Immunol. 2014;153(1):199–208. doi: 10.1016/j.clim.2014.04.012.
    1. Bocchino V, Bertorelli G, Bertrand CP, et al. Eotaxin and Ccr3 are up-regulated in exacerbations of chronic bronchitis. Allergy. 2002;57(1):17–22.
    1. D’Armiento JM, Scharf SM, Roth MD, et al. Eosinophil and T cell markers predict functional decline in COPD patients. Respir Res. 2009;10:113. doi: 10.1186/1465-9921-10-113.
    1. Bade G, Khan MA, Srivastava AK, et al. Serum cytokine profiling and enrichment analysis reveal the involvement of immunological and inflammatory pathways in stable patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2014;9:759–773.
    1. Liu X, Jones GW, Choy EH, Jones SA. The biology behind Interleukin-6 targeted interventions. Curr Opin Rheumatol. 2016;28(2):152–160. doi: 10.1097/BOR.0000000000000255.
    1. Luo Y, Zheng SG. Hall of fame among pro-inflammatory cytokines: interleukin-6 gene and its transcriptional regulation mechanisms. Front Immunol. 2016;7:604. doi: 10.3389/fimmu.2016.00604.
    1. Yende S, Waterer GW, Tolley EA, et al. Inflammatory markers are associated with ventilatory limitation and muscle dysfunction in obstructive lung disease in well functioning elderly subjects. Thorax. 2006;61(1):10–16. doi: 10.1136/thx.2004.034181.
    1. Walter RE, Wilk JB, Larson MG, et al. Systemic inflammation and COPD: the Framingham Heart Study. Chest. 2008;133(1):19–25. doi: 10.1378/chest.07-0058.
    1. Van Durme YM, Lahousse L, Verhamme KM, et al. Mendelian randomization study of Interleukin-6 in chronic obstructive pulmonary disease. Respiration. 2011;82(6):530–538. doi: 10.1159/000332336.
    1. Wei J, Xiong XF, Lin YH, Zheng BX, Cheng DY. Association between serum interleukin-6 concentrations and chronic obstructive pulmonary disease: a systematic review and meta-analysis. PeerJ. 2015;3:E1199. doi: 10.7717/peerj.1199.
    1. Ruwanpura SM, Mcleod L, Miller A, et al. Interleukin-6 promotes pulmonary emphysema associated with apoptosis in mice. Am J Respir Cell Mol Biol. 2011;45(4):720–730. doi: 10.1165/rcmb.2010-0462OC.
    1. Miller J, Edwards LD, Agusti A, et al. Comorbidity, systemic inflammation and outcomes in the eclipse cohort. Respir Med. 2013;107(9):1376–1384. doi: 10.1016/j.rmed.2013.05.001.
    1. Iyer KS, Newell JD, Jr, Jin D, et al. Quantitative dual-energy computed tomography supports a vascular etiology of smoking-induced inflammatory lung disease. Am J Respir Crit Care Med. 2016;193(6):652–661. doi: 10.1164/rccm.201506-1196OC.
    1. Wendling D, Vidon C, Godfrin-Valnet M, Rival G, Guillot X, Prati C. Exacerbation of combined pulmonary fibrosis and emphysema syndrome during Tocilizumab therapy for rheumatoid arthritis. Joint Bone Spine. 2013;80(6):670–671. doi: 10.1016/j.jbspin.2013.03.009.
    1. Knobloch J, Chikosi SJ, Yanik S, Rupp J, Jungck D, Koch A. A systemic defect in toll-liken 4 signaling increases lipopolysaccharide-induced suppression of Il-2-dependent T-cell proliferation in COPD. Am J Physiol Lung Cell Mol Physiol. 2016;310(1):L24–L39. doi: 10.1152/ajplung.00367.2014.
    1. Celik H, Akpinar S, Karabulut H, et al. Evaluation of Il-8 nasal lavage levels and the effects of nasal involvement on disease severity in patients with stable chronic obstructive pulmonary disease. Inflammation. 2015;38(2):616–622. doi: 10.1007/s10753-014-9968-0.
    1. De-Torres JP, Blanco D, Alcaide AB, et al. Smokers with ct detected emphysema and no airway obstruction have decreased plasma levels of Egf, Il-15, Il-8 and Il-1ra. PLoS One. 2013;8(4):E60260. doi: 10.1371/journal.pone.0060260.
    1. Zhang L, Cheng Z, Liu W, Wu K. Expression of interleukin (Il)-10, Il-17a and Il-22 in serum and sputum of stable chronic obstructive pulmonary disease patients. COPD. 2013;10(4):459–465. doi: 10.3109/15412555.2013.770456.
    1. Ying S, O’Connor B, Ratoff J, et al. Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J Immunol. 2008;181(4):2790–2798. doi: 10.4049/jimmunol.181.4.2790.
    1. Saeki H, Tamaki K. Thymus and activation regulated chemokine (Tarc)/Ccl17 and skin diseases. J Dermatol Sci. 2006;43(2):75–84. doi: 10.1016/j.jdermsci.2006.06.002.
    1. Hnatkova M, Mocikova H, Trneny M, Zivny J. The biological environment of Hodgkin’s lymphoma and the role of the chemokine Ccl17/Tarc. Prague Med Rep. 2009;110(1):35–41.

Source: PubMed

3
Sottoscrivi