A blood and bronchoalveolar lavage protein signature of rapid FEV1 decline in smoking-associated COPD

Katarina M DiLillo, Katy C Norman, Christine M Freeman, Stephanie A Christenson, Neil E Alexis, Wayne H Anderson, Igor Z Barjaktarevic, R Graham Barr, Alejandro P Comellas, Eugene R Bleecker, Richard C Boucher, David J Couper, Gerard J Criner, Claire M Doerschuk, J Michael Wells, MeiLan K Han, Eric A Hoffman, Nadia N Hansel, Annette T Hastie, Robert J Kaner, Jerry A Krishnan, Wassim W Labaki, Fernando J Martinez, Deborah A Meyers, Wanda K O'Neal, Victor E Ortega, Robert Paine 3rd, Stephen P Peters, Prescott G Woodruff, Christopher B Cooper, Russell P Bowler, Jeffrey L Curtis, Kelly B Arnold, SPIROMICS investigators, Katarina M DiLillo, Katy C Norman, Christine M Freeman, Stephanie A Christenson, Neil E Alexis, Wayne H Anderson, Igor Z Barjaktarevic, R Graham Barr, Alejandro P Comellas, Eugene R Bleecker, Richard C Boucher, David J Couper, Gerard J Criner, Claire M Doerschuk, J Michael Wells, MeiLan K Han, Eric A Hoffman, Nadia N Hansel, Annette T Hastie, Robert J Kaner, Jerry A Krishnan, Wassim W Labaki, Fernando J Martinez, Deborah A Meyers, Wanda K O'Neal, Victor E Ortega, Robert Paine 3rd, Stephen P Peters, Prescott G Woodruff, Christopher B Cooper, Russell P Bowler, Jeffrey L Curtis, Kelly B Arnold, SPIROMICS investigators

Abstract

Accelerated progression of chronic obstructive pulmonary disease (COPD) is associated with increased risks of hospitalization and death. Prognostic insights into mechanisms and markers of progression could facilitate development of disease-modifying therapies. Although individual biomarkers exhibit some predictive value, performance is modest and their univariate nature limits network-level insights. To overcome these limitations and gain insights into early pathways associated with rapid progression, we measured 1305 peripheral blood and 48 bronchoalveolar lavage proteins in individuals with COPD [n = 45, mean initial forced expiratory volume in one second (FEV1) 75.6 ± 17.4% predicted]. We applied a data-driven analysis pipeline, which enabled identification of protein signatures that predicted individuals at-risk for accelerated lung function decline (FEV1 decline ≥ 70 mL/year) ~ 6 years later, with high accuracy. Progression signatures suggested that early dysregulation in elements of the complement cascade is associated with accelerated decline. Our results propose potential biomarkers and early aberrant signaling mechanisms driving rapid progression in COPD.

Conflict of interest statement

KMD, KCN, CMF, SAC, NEA, NEA, WHA, IZB, RGB, APC, EB, RCB, CBC, DJC, GJC, CMD, JMW, EAH, NNH, ATH, RJK, JAK, DAM, WKO, VEO, RP, SPP, PGW, RPB, JLC, KBA have no competing interests to declare. WWL reports personal fees from Konica Minolta and Continuing Education Alliance. MKH reports personal fees from GlaxoSmithKline, AstraZeneca, Boehringer Ingelheim, Cipla, Chiesi, Novartis, Pulmonx, Teva, Verona, Merck, Mylan, Sanofi, DevPro, Aerogen, Polarian, Regeneron, United Therapeutics, UpToDate, Medscape and Integrity. She has received either in kind research support or funds paid to the institution from the NIH, Novartis, Sunovion, Nuvaira, Sanofi, Astrazeneca, Boehringer Ingelheim, Gala Therapeutics, Biodesix, the COPD Foundation and the American Lung Association. She has participated in Data Safety Monitoring Boards for Novartis and Medtronic with funds paid to the institution. She has received stock options from Meissa Vaccines. FJM. has received personal fees from Forest, Janssen, GlaxoSmithKline, Nycomed/Takeda, Amgen, AstraZeneca, Boehringer Ingelheim, Ikaria/ Bellerophon, Genentech, Novartis, Pearl, Pfizer, Roche, Sunovion, Theravance, Axon, CME Incite, California Society for Allergy and Immunology, Annenberg, Integritas, InThough, Miller Medical, National Association for Continuing Education, Paradigm, Peer Voice, UpToDate, Haymarket Communications, Western Society of Allergy and Immunology, Informa, Bioscale, Unity Biotechnology, ConCert, Lucid, Methodist Hospital, Prime, WebMD, Bayer, Ikaria, Kadmon, Vercyte, American Thoracic Society, Academic CME, Falco, Axon Communication, Johnson & Johnson, Clarion, Continuing Education, Potomac, Afferent, and Adept; and has collected nonfinancial support from Boehringer Ingelheim, Centocor, Gilead, and Biogen/Stromedix; and declares other interests with Mereo, Boehringering Ingelheim, and Centocor. All are outside of this project.

© 2023. The Author(s).

Figures

Figure 1
Figure 1
Individual blood and BAL proteins cannot discriminate between annualized greater versus lesser rates of FEV1 decline in COPD. (a) Comparison of annualized post-bronchodilator FEV1 decline from V1 to V5. Decline was calculated as (V5 FEV1 – V1 FEV1)/ time, where time is the duration in years between V1 and V5 for each participant. (b) Volcano plot of blood and BAL proteins. Light and dark blue protein markers have a p-value < 0.05 and < 0.01, respectively, after a two-sampled two-tailed t-test. All depicted p-values are before correction for multiple comparisons. No proteins remained significant after applying the Benjamini-Hochberg false discovery rate (FDR) correction for multiple comparisons (α = 0.05).
Figure 2
Figure 2
A 52-feature Elastic Net (EN) signature identified individuals at-risk for FEV1 decline ≥70 mL/year with high accuracy. (a) PLSDA scores plot highlighting strong differentiation between greater decliners (magenta) and lesser decliners (yellow), separating the two groups with 98.4% cross-validation (CV) and calibration accuracy. (b) Loadings on latent variable 1 (LV1) (with negatively loaded proteins being comparatively increased in greater decliners and positively loaded proteins being comparatively reduced) captured 11.9% of the total variance in the data. (c) ROC curve of 52-feature signature suggests greater decliners classification with 100% sensitivity and 96.8% specificity in the cross-validated model. (d) LV1 scores were associated with annualized FEV1 decline (mL/yr). P-values and fit line shown for linear models adjusted for age, race, height, sex, baseline FEV1% predicted, smoking status, pack-years, and inhaled corticosteroids (ICS) use within three months of baseline visit. (e,f) Comparison of (e) 6-fold CV accuracies, (f) sensitivities, and specificities between the 52-feature EN signature, a collection of the six top proteins identified in Fig. 1, and literature-based models. All reported values are from cross-validated PLSDA models, unless otherwise noted. One-way ANOVA with Dunnett’s post hoc test; **p<0.01, ****p < 0.0001.
Figure 3
Figure 3
Clustering of COPD subjects by the EN-identified signature highlights distinct regulation of immune-associated processes. (a) Hierarchical clustering of the 52-feature signature highlights distinct clustering of greater decliners (magenta) and lesser decliners (yellow). Only 5 out of the 45 subjects were misclassified (Sensitivity: 85.7%, Specificity: 90.3%). BAL proteins denoted by blue text. (b) Significantly enriched ontology clusters by Metascape analysis. (c, d) Pathways encompassed in the (c) complement system cluster and in the (d) positive regulation of cytokine production cluster are listed in the table. Hatched squares indicate protein involvement in a particular pathway, colorations of magenta or yellow represent a relative elevation of the protein concentration in greater decliners or lesser decliners, respectively.
Figure 4
Figure 4
Complement profiles in COPD lesser decliners behave more similarly to TEPPS than COPD greater decliners. (a) Scores plot from PCA completed using all complement proteins measured in plasma (C1q, C1qBP, C1r, C2, C3d, C3b, C3, C3a, iC3b, C3a des Arg, C4, C4b, C5, C5a, C5-6, C6, C7, C8, C9, Factor B, Factor D, Properdin) of greater decliners (circles), lesser decliners (squares), and a reference group of tobacco-exposed people with preserved spirometry (TEPPS) (diamonds). First two principal components (PCs) capture 33.9% of the variance in the dataset. (b) Comparison of scores on PC1 (one-way ANOVA with Tukey’s post-hoc test; **p<0.01, ***p<0.001).
Figure 5
Figure 5
Select subgroup of signature proteins retain high predictive value for accelerated FEV1 decline. (a) Table denote AUCs for both ROC curves generated from calibration and cross-validation PLSDA models. AUCs of all calibration models were compared to that of the optimal (52-feature model) using the Hanley and McNeil method. (b) Comparisons of 6-fold CV accuracies of multi-compartment and (c) blood-only biomarker models to a collection of the 6 top proteins identified in Fig. 1, and literature models, as determined by ANOVA with Bonferroni’s post hoc test (*p<0.05, **p < 0.01, ***p<0.001). (d) Sensitivity and (e) specificity of signatures. All reported values are from cross-validated PLSDA models, unless otherwise noted. (†: multi-compartment model, #: blood-only model). CV: cross-validated; AUC: area under curve.

References

    1. Kochanek, K. D., Xu, J. & Arias, E. Mortality in the United States, 2019. NCHS Data Brief, no 395 (2020).
    1. Akinbami, L. J. & Liu, X. Chronic obstructive pulmonary disease among adults aged 18 and over in the United States, 1998–2009. NCHS Data Brief, no 63 (2011).
    1. Ford ES, et al. Total and state-specific medical and absenteeism costs of COPD among adults aged ≥ 18 years in the United States for 2010 and projections through 2020. Chest. 2015;147:31–45. doi: 10.1378/chest.14-0972.
    1. Casanova C, et al. The progression of chronic obstructive pulmonary disease is heterogeneous: The experience of the BODE cohort. Am. J. Respir. Crit. Care Med. 2011;184:1015–1021. doi: 10.1164/rccm.201105-0831OC.
    1. Nishimura M, et al. Annual change in pulmonary function and clinical phenotype in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2012;185:44–52. doi: 10.1164/rccm.201106-0992OC.
    1. Mannino DM, Reichert MM, Davis KJ. Lung function decline and outcomes in an adult population. Am. J. Respir. Crit. Care Med. 2006;173:985–990. doi: 10.1164/rccm.200508-1344OC.
    1. Vestbo J, et al. Changes in forced expiratory volume in 1 second over time in COPD. N. Engl. J. Med. 2011;365:1184–1192. doi: 10.1056/NEJMoa1105482.
    1. Zemans, R. L. et al. Multiple biomarkers predict disease severity, progression and mortality in COPD. Respir. Res.18, (2017).
    1. Park HY, et al. Club cell protein 16 and disease progression in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2013;188:1413–1419. doi: 10.1164/rccm.201305-0892OC.
    1. Higashimoto Y, et al. Serum biomarkers as predictors of lung function decline in chronic obstructive pulmonary disease. Respir. Med. 2009;103:1231–1238. doi: 10.1016/j.rmed.2009.01.021.
    1. Bradford E, et al. The value of blood cytokines and chemokines in assessing COPD. Respir. Res. 2017;18:180. doi: 10.1186/s12931-017-0662-2.
    1. Celli BR, et al. Serum biomarkers and outcomes in patients with moderate COPD: A substudy of the randomised SUMMIT trial. BMJ Open Respir. Res. 2019;6:e000431. doi: 10.1136/bmjresp-2019-000431.
    1. Ahmadi-Abhari S, Kaptoge S, Luben RN, Wareham NJ, Khaw KT. Longitudinal association of C-reactive protein and lung function over 13 years:&nbsp;The EPIC-Norfolk study. Am. J. Epidemiol. 2014;179:48–56. doi: 10.1093/aje/kwt208.
    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;62:515–520. doi: 10.1136/thx.2006.066969.
    1. Suzuki M, et al. Lower leptin/adiponectin ratio and risk of rapid lung function decline in chronic obstructive pulmonary disease. Ann. Am. Thorac. Soc. 2014;11:1511–1519. doi: 10.1513/AnnalsATS.201408-351OC.
    1. Liu Z, et al. Identification of potential COPD genes based on multi-omics data at the functional level. Mol. Biosyst. 2016;12:191–204. doi: 10.1039/C5MB00577A.
    1. Zhao P, et al. Integration of transcriptomics, proteomics, metabolomics and systems pharmacology data to reveal the therapeutic mechanism underlying Chinese herbal Bufei Yishen formula for the treatment of chronic obstructive pulmonary disease. Mol. Med. Rep. 2018;17:5247–5257.
    1. Li CX, Wheelock CE, Sköld CM, Wheelock ÅM. Integration of multi-omics datasets enables molecular classification of COPD. Eur. Respir. J. 2018;51:1701930. doi: 10.1183/13993003.01930-2017.
    1. Stockley RA, Halpin DMG, Celli BR, Singh D. Chronic obstructive pulmonary disease biomarkers and their interpretation. Am. J. Respir. Crit. Care Med. 2019;199:1195–1204. doi: 10.1164/rccm.201810-1860SO.
    1. Mannino DM. Biomarkers for chronic obstructive pulmonary disease diagnosis and progression: Insights, disappointments and promise. Curr. Opin. Pulm. Med. 2019;25:144–149. doi: 10.1097/MCP.0000000000000549.
    1. Norman KC, et al. Identification of a unique temporal signature in blood and BAL associated with IPF progression. Sci. Rep. 2020;10:12049. doi: 10.1038/s41598-020-67956-w.
    1. O’Dwyer DN, et al. The peripheral blood proteome signature of idiopathic pulmonary fibrosis is distinct from normal and is associated with novel immunological processes. Sci. Rep. 2017;7:46560. doi: 10.1038/srep46560.
    1. Norman KC, et al. Inference of cellular immune environments in sputum and peripheral blood associated with acute exacerbations of COPD. Cell. Mol. Bioeng. 2019;12:165–177. doi: 10.1007/s12195-019-00567-2.
    1. Wells JM, et al. Safety and tolerability of comprehensive research bronchoscopy in chronic obstructive pulmonary disease. Results from the SPIROMICS bronchoscopy substudy. Ann. Am. Thorac. Soc. 2019;16:439–446. doi: 10.1513/AnnalsATS.201807-441OC.
    1. Freeman CM, et al. Design of a multi-center immunophenotyping analysis of peripheral blood, sputum and bronchoalveolar lavage fluid in the Subpopulations and Intermediate Outcome Measures in COPD Study (SPIROMICS). J. Transl. Med. 2015;13:19. doi: 10.1186/s12967-014-0374-z.
    1. Couper D, et al. Design of the subpopulations and intermediate outcomes in COPD study (SPIROMICS) Thorax. 2014;69:491–494. doi: 10.1136/thoraxjnl-2013-203897.
    1. Perez-Padilla R, et al. Lung function decline in subjects with and without COPD in a population-based cohort in Latin-America. PLoS ONE. 2017;12:e0177032. doi: 10.1371/journal.pone.0177032.
    1. Anderson, W. et al. Classification of lung function trajectories in SPIROMICS [abstract]. Eur. Respir. J.58, Suppl 65, PA1858 (2021).
    1. Regan EA, et al. Omics and the search for blood biomarkers in chronic obstructive pulmonary disease. Am. J. Respir. Cell Mol. Biol. 2019;61:143–149. doi: 10.1165/rcmb.2018-0245PS.
    1. Barnes PJ. Inflammatory mechanisms in patients with chronic obstructive pulmonary disease. J. Allergy Clin. Immunol. 2016;138:16–27. doi: 10.1016/j.jaci.2016.05.011.
    1. Oudijk EJD, Lammers JWJ, Koenderman L. Systemic inflammation in chronic obstructive pulmonary disease. Eur. Respir. Journal, Suppl. 2003;22:5s–13s. doi: 10.1183/09031936.03.00004603a.
    1. Martinez FJ, et al. At the root: Defining and halting progression of early chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2018;197:1540–1551. doi: 10.1164/rccm.201710-2028PP.
    1. Miller RD, Kueppers F, Offord KP. Serum concentrations of C3 and C4 of the complement system in patients with chronic obstructive pulmonary disease. J. Lab. Clin. Med. 1980;95:266–271.
    1. Chauhan S, Gupta MK, Goyal A, Dasgupta DJ. Alterations in immunogolublin & complement levels in chronic obstructive pulmonary disease. Indian J. Med. Res. 1990;92:241–245.
    1. Kosmas EN, Zorpidou D, Vassilareas V, Roussou T, Michaelides S. Decreased C4 complement component serum levels correlate with the degree of emphysema in patients with chronic bronchitis. Chest. 1997;112:341–347. doi: 10.1378/chest.112.2.341.
    1. Baralla A, et al. Plasma proteomic signatures in early chronic obstructive pulmonary disease. Proteomics. Clin. Appl. 2018;12:e1700088. doi: 10.1002/prca.201700088.
    1. Marc MM, et al. Complement factor C5a in acute exacerbation of chronic obstructive pulmonary disease. Scand. J. Immunol. 2010;71:386–391. doi: 10.1111/j.1365-3083.2010.02385.x.
    1. Tan DBA, et al. Protein network analysis identifies changes in the level of proteins involved in platelet degranulation, proteolysis and cholesterol metabolism pathways in AECOPD patients. COPD J. Chronic Obstr. Pulm. Dis. 2020;17:29–33. doi: 10.1080/15412555.2019.1711035.
    1. Sun W, et al. Common genetic polymorphisms influence blood biomarker measurements in COPD. PLoS Genet. 2016;12:e1006011. doi: 10.1371/journal.pgen.1006011.
    1. Mastej E, et al. Identifying protein–metabolite networks associated with COPD phenotypes. Metabolites. 2020;10:124. doi: 10.3390/metabo10040124.
    1. Mahesh M, Yalamudi M, Lokesh S. Complement levels in chronic obstructive pulmonary disease: Correlation with pulmonary function and radiological emphysema score. Int. J. Sci. Study. 2016;3:280–284.
    1. Yuan X, et al. Activation of C3a receptor is required in cigarette smoke-mediated emphysema. Mucosal Immunol. 2015;8:874–885. doi: 10.1038/mi.2014.118.
    1. Marc MM, et al. Complement factors C3a, C4a, and C5a in chronic obstructive pulmonary disease and asthma. Am. J. Respir. Cell Mol. Biol. 2004;31:216–219. doi: 10.1165/rcmb.2003-0394OC.
    1. Kulkarni HS, et al. Intracellular C3 protects human airway epithelial cells from stress-associated cell death. Am. J. Respir. Cell Mol. Biol. 2019;60:144–157. doi: 10.1165/rcmb.2017-0405OC.
    1. Sohal SS, et al. Reticular basement membrane fragmentation and potential epithelial mesenchymal transition is exaggerated in the airways of smokers with chronic obstructive pulmonary disease. Respirology. 2010;15:930–938. doi: 10.1111/j.1440-1843.2010.01808.x.
    1. Mahmood MQ, et al. Epithelial mesenchymal transition in smokers: large versus small airways and relation to airflow obstruction. Int. J. Chron. Obstruct. Pulmon. Dis. 2015;10:1515–1524. doi: 10.2147/COPD.S81032.
    1. Milara J, Peiró T, Serrano A, Cortijo J. Epithelial to mesenchymal transition is increased in patients with COPD and induced by cigarette smoke. Thorax. 2013;68:410–420. doi: 10.1136/thoraxjnl-2012-201761.
    1. Nishioka, M. et al. Fibroblast-epithelial cell interactions drive epithelial-mesenchymal transition differently in cells from normal and COPD patients. Respir. Res.16, (2015).
    1. Cockayne DA, et al. Systemic biomarkers of neutrophilic inflammation, tissue injury and repair in COPD patients with differing levels of disease severity. PLoS One. 2012;7:e38629. doi: 10.1371/journal.pone.0038629.
    1. Lai T, et al. Heparin-binding epidermal growth factor contributes to COPD disease severity by modulating airway fibrosis and pulmonary epithelial-mesenchymal transition. Lab. Invest. 2018;98:1159–1169. doi: 10.1038/s41374-018-0049-0.
    1. Wei Y, et al. Prognostic value of cripto-1 expression in non-small-cell lung cancer patients: a systematic review and meta-analysis. Biomark. Med. 2020;14:317–329. doi: 10.2217/bmm-2019-0159.
    1. Nunomiya K, et al. Relationship between serum level of lymphatic vessel endothelial hyaluronan receptor-1 and prognosis in patients with lung cancer. J. Cancer. 2014;5:242–247. doi: 10.7150/jca.8486.
    1. Nishimura M, et al. Annual change in pulmonary function and clinical phenotype in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2012;185:44–52. doi: 10.1164/rccm.201106-0992OC.
    1. Higham A, Quinn AM, Cançado JED, Singh D. The pathology of small airways disease in COPD: historical aspects and future directions. Respir. Res. 2019;20:49. doi: 10.1186/s12931-019-1017-y.
    1. Raffield LM, et al. Comparison of proteomic assessment methods in multiple cohort studies. Proteomics. 2020;20:e1900278. doi: 10.1002/pmic.201900278.
    1. O’Neal, W. K. et al. Comparison of serum, EDTA plasma and P100 plasma for luminex-based biomarker multiplex assays in patients with chronic obstructive pulmonary disease in the SPIROMICS study. J. Transl. Med.12, (2014).
    1. Gold L, et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE. 2010;5:e15004. doi: 10.1371/journal.pone.0015004.
    1. Hanley JA, McNeil BJ. A method of comparing the areas under receiver operating characteristic curves derived from the same cases. Radiology. 1983;148:839–843. doi: 10.1148/radiology.148.3.6878708.
    1. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun.10, (2019).
    1. Qian, J., Hastie, T., Friedman, J., Tibshirani, R. & Simon, N. Glmnet for MATLAB. (2013).

Source: PubMed

3
订阅