Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2017

Open Access 01.12.2017 | Research article

Biomarkers of collagen turnover are related to annual change in FEV1 in patients with chronic obstructive pulmonary disease within the ECLIPSE study

verfasst von: Diana J. Leeming, Inger Byrjalsen, Jannie M. B. Sand, Asger R. Bihlet, Peter Lange, Ruth Thal-Singer, Bruce E. Miller, Morten A. Karsdal, Jørgen Vestbo, The Evaluation of COPD Longitudinally to Identify Surrogate Endpoints (ECLIPSE) study investigators

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2017

Abstract

Background

Change in forced expiratory volume in one second (FEV1) is important for defining severity of chronic obstructive pulmonary disease (COPD). Serological neoepitope markers of collagen turnover may predict rate of change in FEV1.

Methods

One thousand COPD subjects from the observational, multicentre, three-year ECLIPSE (Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints) study (NCT00292552, trial registration in February 2006) were included. Matrix metalloproteinase (MMP)-generated fragments of collagen type I, and type VI (C1M and C6M) were assessed in month six serum samples. A random-coefficient model with both a random intercept and a random slope was used to test the ability of the markers to predict post-dose bronchodilator FEV1 (PD-FEV1) change over two years adjusting for sex, age, BMI, smoking, bronchodilator reversibility, prior exacerbations, emphysema and chronic bronchitis status at baseline.

Results

Annual change of PD-FEV1 was estimated from a linear model for the two-year study period. Serum C1M and C6M were independent predictors of lung function change (p = 0.007/0.005). Smoking, bronchodilator reversibility, plasma hsCRP and emphysema were also significant predictors. The effect estimate between annual change in PD-FEV1 per one standard deviation (1SD) increase of C1M and C6M was +10.4 mL/yr. and +8.6 mL/yr. C1M, and C6M, had a significant association with baseline FEV1.

Conclusion

We demonstrated that markers of tissue turnover were significantly associated with lung function change. These markers may function as prognostic biomarkers and possibly as efficacy biomarkers in clinical trials focusing on lung function change in COPD.

Trial registration

NCT00292552, Retrospectively registered, trial registration in February 2006.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12890-017-0505-4) contains supplementary material, which is available to authorized users.
Abkürzungen
%LAA
Percentage of lung CT voxels below a threshold of −950 Hounsfield units and percent low areas of attenuation
1SD
One standard deviation
ATS-DLD
American thoracic society–division of lung disease
BL
Baseline
BODE
Body-mass index, airflow obstruction, dyspnoea, and exercise index
C1M
Marker of MMP degraded type I collagen
C6M
Marker of MMP degraded type VI collagen
CC-16
Club Cell protein-16
COPD
Chronic obstructive pulmonary disease (COPD)
CT
Computed tomographic
ECLIPSE
Evaluation of COPD longitudinally to identify predictive surrogate endpoints study ELISA
Enzyme linked immunosorbent assay
ECM
Extracellular matrix
FDA
Food and drug administration
FEV1
Forced expiratory volumen in 1 s
FVC
Forced vital capacity)
GOLD
The global initiative for obstructive lung disease
hsCRP
High sensitive C-reactive protein
IPF
Idiopathic pulmonary fibrosis
KL-6
Krebs von den Lungen-6
M6
Month 6
MMP
Matrix metalloproteinase
PD-FEV1
Post-dose bronchodilator FEV1
SP-A
Surfactant protein-A
SP-D
Surfactant protein-D
WCB
White blood cells

Background

Chronic obstructive pulmonary disease (COPD) is a leading cause of morbidity and mortality worldwide and is a disease leading to narrowing of the small airways, airway fibrosis and chronic changes in the lung parenchyma [1] mainly due to cigarette- or biomass smoke exposure [2]. Phenotyping of patients is generally based on a combination of clinical and morphological features such as frequency of exacerbations, lung function, presence or absence of emphysema and chronic bronchitis, and presence of asthmatic features [3, 4]. Therapies affecting such parameters in COPD are limited [5].
In COPD, the airway composition and structure is changed compared to the healthy state, due to small airway inflammation resulting in epithelial damage and subsequent airway fibrosis with increased deposition and remodelling of extracellular matrix (ECM) proteins in the basement membrane and interstitial matrix [68] of the lung. Activated lung fibroblasts deposit ECM proteins, such as collagens type I and III, fibronectin, laminin [7, 911], biglycan, decorin and perlecan [12], all alleged of having severe effects on the lung integrity. These proteins are believed to be elevated and remodelled during chronic inflammation in the small airways. Furthermore, pathological proteases such as elastase [13], and matrix metalloproteinase (MMP)-1, −2, −7, −9 and −12 [14] have been reported to be over-expressed in COPD affected tissue [15]. The most abundant collagen in the interstitial matrix of the lung is the fibril-forming type I collagen [16] which is mainly found located alongside with type III collagen, the second most abundant collagen type. Together they provide the structural framework of the alveolar wall, pulmonary blood vessels, visceral pleura and the connective tissue sheaths that surround the tracheobronchial tree [17, 18]. Type VI collagen is another important member of the collagens of the interstitial matrix, which main function is to connect cells to territorial matrix [19, 20]. Recently, it was shown that circulating neoepitope markers of ECM were related to clinical outcomes in patients with idiopathic pulmonary fibrosis (IPF) [21]. In IPF, serum markers of MMP-generated fragments of type I, III, VI collagen, biglycan and CRP were able to identify patients with fast progression and high risk of mortality. Generally, markers of ECM remodellings including C1M and C6M have been shown to relate to disease activity in the osteoporosis, COPD and pagets disease, rheumatoid arthritis (RA), hepatitis C, hepatitis B [2231] C6M and C1M has been shown to be elevated in COPD compared to healthy individuals; C6M is upregulated during AECOPD, however not related to smoking status or pack years [3235]. In the present work, we aimed to investigate the ability of novel serological ECM related neoepitope markers to predict rate of change of forced expiratory volume in one second (FEV1) in COPD patients within the ECLIPSE study [36]. The markers utilized are highly specific for the detection of MMP-mediated fragments of type I (C1M [37]) or type VI collagen (C6M [19]). The hypothesis was that some of these ECM markers, which are markers of pulmonary fibrosis in patient with idiopathic pulmonary fibrosis (IPF) [21], were able to predict lung function change in patients with pulmonary structural changes due to COPD. We speculated that these markers may be valid as an aid in the selection of patients in most need of treatment, and patients for clinical trials focusing on progression of COPD.

Methods

Study design

The present analysis was performed in the three-year ECLIPSE observational study (ClinicalTrials.​gov number, NCT00292552, trial registration in February 2006) described previously [36, 38]. The full ECLIPSE study included 2164 patients with COPD. Briefly, patients included were between 40 and 75 years at baseline, had a history of ten or more pack-years of smoking, a post-dose bronchodilator FEV1 less than 80% of the predicted value and a post-dose bronchodilator ratio of FEV1 to forced vital capacity (FVC) of 0.7 or less. The Global initiative for Obstructive Lung Disease (GOLD) spirometry stages included were II-IV. Respiratory symptoms, smoking history, occupational exposure, and coexisting medical conditions were documented at study entry with the use of a modified version of the American Thoracic Society–Division of Lung Disease (ATS-DLD) questionnaire. The study was conducted in accordance with the Declaration of Helsinki and good clinical practice guidelines, and was approved by all local and participating centres’ ethics and review boards reference number SCO104960 (ECLIPSE) RM 2005/00273/05 (Additional file 1). All patients provided their informed written consent. A list of the particpating centers is seen in Additional file 1. Each patient underwent spirometry before and 15 min after inhaling 400 μg of salbutamol (GlaxoSmithKline). Computed tomographic (CT) scanning of the chest was performed at baseline to evaluate the severity and distribution of emphysema. Quantitative assessment of lung volumes and estimation of the percentage of lung CT voxels below a threshold of −950 Hounsfield units and percent low areas of attenuation (%LAA) was performed using Pulmonary Workstation software, version 2.0 (VIDA Diagnostics). Significant amount of emphysema was defined as having more than 10%LAA. Whole blood was collected by venipuncture into vacutainer tubes. Serum was prepared by allowing the blood to clot for 30 min at room temperature followed by centrifugation at 1500 g for 10–15 min. Plasma was obtained by centrifugation of vacutainer tubes at 2000 g for 10–15 min.

Definition of subpopulation

A subpopulation of 1000 patients was selected from the ECLIPSE cohort for the purpose of evaluating the ability of two serum markers of ECM remodeling to predict the longitudinal change in FEV1 within the study compared to existing biomarkers assessed within the study. The 500 most rapid post-dose bronchodilator FEV1 (PD-FEV1) decliners and the 500 slowest PD-FEV1 decliners were selected for the biomarker assessments. The linear PD-FEV1 decline was defined as the change in PD-FEV1 from year 1 (Y1) to year 3 (Y3). Baseline to month 6 (M6) PD-FEV1 data were excluded in the present analysis since the biochemical markers included in the present study analysis were assessed in serological samples at different time points: baseline (BL), M6 or Y1. A schematic overview of the data used is seen in Fig. 1. This way only PD-FEV1 data obtained post blood sampling were used in the analysis.

Quantification of biochemical markers in the subpopulation

Serum samples from the M6 visit that had been stored at −80 °C until used for analysis. Serum levels of a MMP derived collagen type I fragment (C1M) [37] and a MMP derived collagen type VI fragment (C6M) [19] were assessed using competitive enzyme linked immunosorbent assays (ELISAs) (Nordic Bioscience, Herlev, DK) in the subpopulation of 1000 patients. Technical data on robustness, specificity and analyte stability are published for C1M and C6M [37, 19]. Biomarkers were assessed in a blinded manner according to the manufacturer and all samples were measured within the detection range. EDTA plasma levels of CC-16 at BL, hsCRP and fibrinogen at Y1 had prior to this study been assessed in the total ECLIPSE cohort [39]. Thus data of CC-16, hsCRP and fibrinogen from the ECLIPSE database were used for analysis.

Statistical analysis

Demographic characteristics have been summarized as mean with standard deviation (SD) or number of subjects with percentage as applicable. The change in FEV1 over time was estimated from five FEV1 measurements performed through Y1 to Y3 in the study using a linear regression model. Negative PD-FEV1 refers to a loss of FEV1 and positive PD-FEV1 refers to a retaining of PD-FEV1. A saddle test was performed (proc modeclus) on all FEV1 change data using a radius of the sphere of support for uniform-kernel density estimation and the neighborhood for clustering of R = 10 or R = 20. The association of PD-FEV1 and PD-FEV1 in percent of predicted value (PD-FEV1%) was calculated using a random-coefficient model with both a random intercept and a random slope including the co-variates gender, age, BMI, current smoking status, bronchodilator reversibility, number of prior acute exacerbations, emphysema, chronic bronchitis status, and number of pack-years at baseline. A multimarker random-coefficient model including C1M, C6M, fibrinogen, and hsCRP in one model or using stepwise exclusion tested for association to the linear change of PD-FEV1 was performed. Furthermore, the association to the linear change of PD-FEV1 was calculated for the total population; in patients stratified according to GOLD stages B, C and D; and in patients defined as slow or rapid decliners using a cut-off value of FEV1 of 40 mL/yr. Levels of C1M, C6M and hsCRP in patients with or without significant emphysema (%LAA < 10) were tested using a Mann-Whitney unpaired t-test. The SAS software package (release 9.3; SAS Institute Inc., Cary, NC, USA) was used for the statistical calculations.

Funding source

The study was sponsored by GlaxoSmithKline; the Danish Agency for Science, Technology and Innovation; and the Danish Research Foundation. Two representatives of GlaxoSmithKline and one academic, together representing the ECLIPSE study investigators, two representatives of Nordic Bioscience developed the present study design and concept. All approved the plan for the current analyses, had full access to the data, and were responsible for the decision to publish. The study sponsor did not place any restrictions with regard to statements made in the final paper.

Results

The demographic description of the patient population is presented in Table 1. The mean annual rates of change in PD-FEV1 and PD-FEV1% in all patients from year 1 to year 3 were −43 ± 120 mL and −1.5 ± 4.2%, respectively. The mean annual rates of change were −132 ± 91 mL and −4.7 ± 3% for PD-FEV1 and PD-FEV1% in the 500 most rapid decliners and +44 ± 95 mL and +1.6 ± 3% in the slowest decliners. The distribution of estimated FEV1 change (Fig. 2) showed that most patients lost between −20 and −200 mL/yr. (60%) or gained 20–160 mL/yr. (30%) in FEV1 in the two-year period. A saddle test of the estimated FEV1 change distribution did not indicate that this parameter followed a bi-phasic distribution. Patients in the population did genereally have a stable annual rate of change in FEV1. C1M, C6M and hsCRP were not significantly elevated in patients with significant emphysema (%LAA > 10) compared to those without.
Table 1
Population demographics at baseline
Variable
N
Mean
SD
SEM
Age (yr)
1000
63.1
7.21
0.2
BMI
1000
26.9
5.9
0.2
Gender (F/M)
363/637
GOLD stage
II(494), III(406), IV(100)
PD-FEV1%
998
50.6
15.2
0.5
%LAA
887
16.3
11.3
0.4
F Female, M Male, PD-FEV 1 % Percent predicted normal post dose forced expiratory volume in 1 s, %LAA Percent low areas of attenuation below −950 Hounsfield units, BMI Body mass index

Association between clinical patient characteristics or single biomarkers and baseline FEV1

There was a statistical significant association between a range of patient characteristics or biomarkers and baseline PD-FEV1 (Table 2). Age, BMI, sex, emphysema, bronchodilator reversibility and no prior exacerbation showed the most pronounced association (p < 0.0001) in the total population. An overview of the association for significant parameters is seen in Fig. 3, showing that both patient characteristics and biomarkers were able to describe baseline FEV1 difference. The association of a 1SD change in a biomarker was calculated for each marker in the subpopulation for PD-FEV1 showing associations between −53.3 to 38.1 mL (Table 2). Plasma fibrinogen and hsCRP had the largest association (−53.3/−53.3 mL, p = 0.0002/0.0003), and serum C1M and C6M also showed significant association (−45.2 mL, p = 0.002; −46.5 mL, p = 0.0011) to baseline FEV1.
Table 2
Association between patients’ characteristics and biomarkers on baseline Forced Expiratory Volume in 1 Second (FEV1) assessed in millilitres (mL) and annual rate of change in FEV1. Data are shown as mean ± standard error (SE). Data are generated from a random-coefficient model. The p-values are not corrected for multiple testing
Marker
Associationa with baseline FEV1
P value
Associationa with annual rate of change in FEV1
P value
mL
mL/yr
Serological biomarkers
 BL CC-16
38.1 ± 15.2
0.012
−3.0 ± 4.0
0.50
 BL SP-D
7.6 ± 14.4
0.60
−2.9 ± 3.8
0.50
 BL WBC
−24.0 ± 14
0.09
−2.8 ± 3.8
0.50
 MO6 C1M
−45.2 ± 14.4
0.002
10.4 ± 3.9
0.007
 MO6 C6M
−46.5 ± 14.2
0.0011
8.6 ± 3.8
0.03
 Y1 hsCRP
−53.3 ± 14.7
0.0003
8.2 ± 4.0
0.04
 Y1 Fibrinogen
−53.3 ± 14.3
0.0002
6.8 ± 3.9
0.08
Patients characteristics at baseline
 Age (per yr)
−13.1 ± 2.1
< 0.0001
0.9 ± 0.6
0.10
 SEX (female vs male)
−358 ± 30.9
< 0.0001
6.8 ± 8.2
0.41
 BMI (kg/m2)
10.7 ± 2.7
< 0.0001
0.2 ± 0.7
0.79
# prior AECOPD
 1 vs 0
−172 ± 35.0
< 0.0001
2.8 ± 9.6
0.77
 2 vs 0
−151 ± 42.8
0.0004
18.9 ± 11.8
0.11
≥ 3 vs 0
−323 ± 42.2
< 0.0001
13.9 ± 13.8
0.31
AECOPD FU
Na
Na
−1.8 ± 1.9
0.35
Current smoker (yes vs no)
−19.4 ± 32.1
0.55
−30.7 ± 8.5
0.0003
Bronchodilator reversibility (yes vs. no)
241 ± 34.3
< 0.0001
−22.8 ± 9.1
0.012
Significant emphysema (yes vs. no)
−347 ± 31.6
< 0.0001
−27.9 ± 8.4
0.001
Chronic bronchitis (yes vs no)
−35.4 ± 31.1
0.26
−9.7 ± 8.2
0.24
#Pack years
−0.8 ± 0.6
0.17
0.1 ± 0.2
0.68
CC-16 Club-cell secretory protein-16, SP-D Surfactant protein-D, WBC white blood cell count, C1M MMP derived collagen type I fragment, C6M MMP derived collagen type VI fragment, hsCRP high sensitive C-reactive protein, BMI Body mass index, AECOPD Acute exacerbations in COPD, FU Follow up, aThe association are calculated as per increase in 1SD in each biomarker or as indicated for each patient characteristics. na: non applicable
all p values below <0.05 should be bold and italic

Association between clinical patient characteristics or single biomarkers and annual FEV1 change

The effect estimate of a 1SD change in a biomarkers was calculated for each marker in the total population for the PD-FEV1 and PD-FEV1% linear change from Y1 to Y3 (Table 2). An overview of the association for significant parameters is seen in Fig. 3, generally showing that patient characteristics and biomarkers were able to describe annual change in FEV1 with patient characteristics providing negative associations and serological markers providing positive associations. Serum C1M and C6M at 6 months were independent predictors of lung function change. A 1SD increase in C1M accounted for a decreased rate of decline of lung function of 10.4 mL/yr. (p = 0.007) in PD-FEV1. For C6M, a 1SD increase accounted for a a decreased rate of annual decline in lung function of 8.6 mL/yr. (p = 0.03) in PD-FEV1. The rate of change in PD-FEV1% for C1M was +0.37% (p = 0.005) and for C6M +0.29% (p = 0.03). The association between C1M and C6M and PD-FEV1 and PD-FEV1% change was not affected by baseline age, sex, GOLD stage, smoking status, emphysema, bronchodilator reversibility, and number of previous exacerbations. Y1 plasma hsCRP accounted for a rate of change in PD-FEV1 of 8.2 mL (p = 0.04) and 0.30% for PD-FEV1% (p = 0.03). Smoking status, bronchodilator reversibility, and emphysema were also confirmed as predictors. Fibrinogen and CC-16 were not able to predict rate of Y1 to Y3 annual decline for PD-FEV1 nor PD-FEV1% in this subpopulation of the ECLIPSE study. The estimated annual loss in FEV1 using the multimarker random-coefficient model for C1M, C6M and hsCRP is shown in Fig. 4. Here the 25th- and the 75th percentile groups are plotted showing the estimated loss in these two groups. For C1M, the annual loss of FEV1 was higher in patients below 46.3 ng/mL (25th percentile group) versus in patients below 105.5 ng/mL (75th percentile group). For C6M and hsCRP, the 25th percentile was 11.7- or 1.5 ng/mL, respectively and the 75th percentile 22.5- or 7.45 ng/mL, respectively. There was no significant associations to PD-FEV1 change within different GOLD stages, or in patients either being slow- or rapid decliners. A multimarker model including C1M, C6M, fibrinogen and hsCRP did not increase the association to linear change in PD-FEV1 compared to the use of single markers.

Discussion

To the best of our knowledge, the present study is the first of its kind to evaluate the ability of novel serological ECM neoepitope markers, reflecting collagen type I and VI degradation, to predict the rate of change in lung function in patients with COPD. We demonstrated that serum C1M and C6M were independent predictors of lung function change in COPD patients over a two year period follow up. Furthermore, a significant correlation at baseline to FEV1 by several markers and patient characteristic was observed within the study.
The average annual rate of change in lung function was evaluated in the ECLIPSE study reporting a FEV1 decline of 33 ml per year, including patients that declined, were stable or improved in FEV1 [36]. Several clinical trials have shown that FEV1 decline was higher in mild COPD compared to more severe disease [4043]. Such observations imply that COPD encloses a variety of patients with diverse FEV1 trajectories [44]. Thus, it has become a challenge to stratify patients into phenotypes such as slow and rapid FEV1 decliners. Better patient stratification and phenotyping by disease activity biomarkers would provide a highly valued tool for patient handling in general practice and in clinical trials to enrich the patient population [45].
Non-serological markers such as the "Body-mass index, airflow Obstruction, Dyspnoea, and Exercise" index (BODE), walking tests, exacerbation rate and dyspnoea score are known to be related to disease progression defined by loss of FEV1 and mortality in patients with COPD [36, 39]. However, there is a medical need for serological markers of progressive COPD and selection of patient phenotypes at baseline [46]. It was previously shown in the ECLIPSE (Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints) study [36] that patient characteristics, but not most serological biomarkers, were associated to change in lung function over the three-year period of the study. The analysis of biomarkers included baseline assessed fibrinogen, interleukin-6, interleukin-8, TNF-a, high sensitive C-reactive protein (hsCRP), Club Cell protein-16 (CC-16) and surfactant protein D in a subset of 1793 patients. Only CC-16 was significantly associated with lung function change. Increase of one standard deviation of CC-16 was equal to the protection of 4 ml/yr. (p = 0.04) in lung function change. hsCRP was only borderline significant (p = 0.07), but indicated that increased levels were associated with a lower rate of lung function decline.
Most serological markers evaluated in stable COPD, have shown limited value in relation to the pathology in COPD including sputum neutrophils, circulating white blood cells (WBC), and protein biomarkers [47]. Krebs von den Lungen-6 (KL-6), CC-16, surfactant protein-A (SP-A) and surfactant protein-D (SP-D) are discussed as serological markers of pulmonary diseases [4851]. They have, however, not been convincingly demonstrated to be of value for patient phenotyping in COPD [4851]. Desmosine and isodesmosine, two molecules involved in elastin cross-linking [48], have shown some value [52], but have not been fully validated for use in COPD. A marker of blood clotting, fibrinogen, was recently approved by the Food and Drug Administration (FDA) as a drug development tool for the use of patient enrichment in clinical intervention trials [53]. Fibrinogen appears to be related to COPD exacerbations and mortality and used as an aid in selection of patients that are most likely to progress to a clinical outcome that may be used as a hard endpoint [53].
At present, it has not yet been investigated whether pulmonary structural changes in the lung ECM in COPD is interrelated to rate of change in lung function. In a small number of patients with mild COPD, it has been shown that C1M and C6M were elevated when compared to healthy controls [54]. Furthermore, important data have been presented showing that these ECM neoepitope markers are related to exacerbations [32] and mortality in COPD [55]. A range of serological biomarkers have previously been evaluated in the full ECLIPSE study as predictors of annual rate of change in FEV1, yet only CC-16 was associated with a decreased rate of annual decline in lung function with a modest effect estimate of +4 ± 2 mL/yr. FEV1 per 1 SD change in plasma CC-16 [36, 56]. In the present study, serum C1M, C6M and plasma hsCRP were significantly related to a decreased rate of annual decline in lung function ranging from +8.2 to +10.4 mL/yr. FEV1 per 1SD change in each marker during a two year follow up, which was superior to the association observed for CC-16 [36]. In the subpopulation analysed in the present work, CC-16 was not significantly related to the rate of lung function change. It was notable that increasing levels of all the significant related markers including CC-16 in the full ECLIPSE study [36] were related to a lower rate of lung function decline which may indicate that patients with impaired tissue repair, thus low biomarker levels, progress the fastest in relation to lung function decline. This is however speculative at present. The data did not change when patients were stratified according to GOLD stage C-D or being slow- or rapid lung function decliners. These data were nevertheless unexpected since we intuitively regard elevated levels of ECM remodelling and CRP as being related to poor outcome in patients. In the literature it has been presented that serum hsCRP was prognostic of mortality in stable COPD [57, 58] and that persistent systemic inflammation, here among others plasma CRP, is associated with poor clinical outcome in the ECLIPSE cohort [59]. Nevertheless, persistent inflammation was not related to the prevalence of chronic bronchitis, degree of emphysema, bronchodilator reponse and rate of FEV1 decline in the population investigated [59]. Finally, WBC, C1M, C6M, hsCRP and Fibrinogen were associated with a lower baseline FEV1 indicating that severe patients according to lung function have higher biomarker levels, correlating to the data published on death outcome [55], as previously mentioned. Lange et al. [60] has published how the effect of small lungs from early age has an effect on FEV1, thus an effect on diagnosis of COPD, and potentially COPD phenotypes, and FEV1 change. The ability of predicting annual rate of change in FEV1 may be different in mild COPD vs moderate to severe but also depend on baseline FEV1.
In a small study of COPD patients, Ferrari et al. indicated that plasma hsCRP was not related to mortality [61], thus data are to some extent contradictory. Inconsistently to the data in the present publication, Shabann et al. revealed that the highest tertile of serum CRP levels was associated with greater decline in annual FEV1 in COPD patients [62], which was not the conclusion of the present work. The discrepancy between the two studies may relate to the large differences in age, BMI and FEV1 at study entry. The COPD patients investigated by Shaaban et al. [62] were younger (mean 36.6 yr), more lean (BMI = 22.6) and had a higher FEV1% (105%), however had similar rate of annual decline in lung function (33 vs 29 ml). Our findings indicate that various patohology linked pathways in COPD have a distinct effect on the ECM remodelling thus levels of ECM neo-epitopes. Finally, in the ECLIPSE study analysis, we have observed that C1M, C6M and hsCRP are elevated in patients that died within the study period compared to those who did not {Sand, 2016 4514 /id, which indicates that high levels of such markers are related to a poor outcome.
In the present study, there was no relation between the structure related markers C1M and C6M or CRP with having significant emphysema or not. This may be explained by the fact that the emphysema defined groups are still very heterogeneous and have different ECM remodelling profiles. It appears that assessment of MMP degraded collagen type I or type VI and total CRP are not the correct markers to evaluate emphysema in a broad population.
The stability of C1M and C6M has in a large cohort of patients with IPF been shown over a six month time period [21]. Here it was seen that levels were stable over time and that patients within the high level or low level group at baseline stayed within this group over time. Nevertheless, it was seen that a relative small increase of C1M and C6M over time in individual IPF patients were related to loss of FVC or death {Jenkins, 2015 4101 /id}. These data, eventhough being in IPF, which is a fibrogenesis related disease, are contradictory to the present findings in COPD.
We confirmed in this subpopulation that smoking status, bronchodilator reversibility, and emphysema were significantly related to the annual rate of FEV1-decline ranging between −30.7-27.9 mL/yr. A pronounced and significant association by both serological markers and patient characteristics was seen on baseline FEV1. Serum C1M and C6M, plasma CC-16, hsCRP, and fibrinogen, age, sex, BMI, prior exacerbations, bronchodilator reversibility and emphysema had a significant association with baseline lung capacity. This highlights the importance of these markers and characteristics in the description of COPD traits and potentially their role in patient stratification. The use of a single marker reflecting few pathophysiological traits may not be sufficient whereas a panel of markers most likely may be required for optimal prognosis assessment and evaluation of efficacy of intervention of upcoming drugs. However, neither C1M nor C6M did in the multimarker model increase the association with annual change.

Limitations

In general, the rate of decline in FEV1 observed within the ECLIPSE study was small and highly variable [36] providing a small window of progression during the two year period. Furthermore, FEV1 measurements are generally recognized as being highly variable. In the present study, the two extreme groups in FEV1 change were selected for analysis thus leaving out the patients with FEV1 changes in-between; this may have added to lowering data noise observed in real world COPD studies. Generally, the analysis of serological biochemical markers using the ELISA technique often has limitations such as background noise from the normal level of tissue remodelling and variation inherited into the method. Here the signal-to-noise ratio may be improved by incorporation of disease-related post-translational modifications such as protease cleavage into the specificity criteria of a given assay [63].
In perspective, novel protein fingerprint fibrosis markers as evaluated in the present study may provide a new clinical opportunity for improved evaluation of the involvement of small airways fibrosis activity in COPD. This may add to phenotype stratification and selection of patients for clinical trials. However, this needs to be proven in future prospective studies. Nevertheless, the data at present are preliminary and verification in a second cohort is needed including evaluation in milder COPD. Furthermore, data should additionally be evaluated in a “real world” COPD study where the criteria of extreme annual rate of FEV1 is not included.

Conclusion

In conclusion, we demonstrated that extracellular matrix markers of tissue turnover were predictive of FEV1 change in patients with COPD and suggest that such markers may be used as prognostic biomarkers and possibly biomarkers for patient enrichment in clinical trials.

Acknowledgements

The authors acknowledge all participants, medical, nursing, and technical staff involved in the ECLIPSE study, as well as the technical staff responsible for biomarker assessments.
The Evaluation of COPD Longitudinally to Identify Surrogate Endpoints (ECLIPSE) study principle investigators and centers (NCT00292552, SC0104960) (Additional file 2):
Bulgaria: Y Ivanov, Pleven; K Kostov, Sofia. Canada: J Bourbeau, Montreal; M Fitzgerald, Vancouver; P Hernández, Halifax; K Killian, Hamilton; R Levy, Vancouver; F Maltais, Montreal; D O’Donnell, Kingston. Czech Republic: J Krepelka, Praha. Denmark: J Vestbo, Hvidovre. The Netherlands: E Wouters, Horn. New Zealand: D Quinn, Wellington. Norway: P Bakke, Bergen, Slovenia: M Kosnik, Golnik. Spain: A Agusti, Jaume Sauleda, Palma de Mallorca. Ukraine: Y Feschenko, Kiev; V Gavrisyuk, Kiev; L Yashina, W MacNee, Edinburgh; D Singh, Manchester; J Wedzicha, London. USA: A Anzueto, San Antonio, TX; S Braman, Providence. RI; R Casaburi, Torrance CA; B Celli, Boston, MA; G Giessel, Richmond, VA; M Gotfried, Phoenix, AZ; G Greenwald, Rancho Mirage, CA; N Hanania, Houston, TX; D Mahler, Lebanon, NH; B Make, Denver, CO; S Rennard, Omaha, NE; C Rochester, New Haven, CT; P Scanlon, Rochester, MN; D Schuller, Omaha, NE; F Sciurba, Pittsburg, PA; A Sharafkhaneh, Houston, TX; T Siler, St Charles, MO; E Silverman, Boston, MA; A Wanner, Miami, FL; R Wise, Baltimore, MD; R ZuWallack, Hartford, CT.
Steering Committee: H Coxson (Canada), L Edwards (GlaxoSmithKline, USA), C Crim (GlaxoSmithKline, USA) D Lomas (UK), W MacNee (UK), E Silverman (USA), R Tal-Singer (Co-Chair, GlaxoSmithKline, USA), J Vestbo (Co-chair, Denmark), J Yates (GlaxoSmithKline, USA).
Scientific Committee: A Agusti (Spain), P Calverley (UK), B Celli (USA),), B Miller(GlaxoSmithKline, US), W MacNee (Chair, UK), S Rennard (USA), R Tal-Singer (GlaxoSmithKline, USA), E Wouters (The Netherlands), J Yates (GlaxoSmithKline, USA).

Funding

The study was sponsored by GlaxoSmithKline; the Danish Agency for Science, Technology and Innovation; and the Danish Research Foundation. The funding bodies did not play a role in the design of the study, or collection, analysis, and interpretation of data, or in writing the manuscript.

Availability of data and materials

The datasets generated and/or analysed during the current study are at the moment not publicly available due that data are owned by GSK for the ECLIPSE study and the ECM biomarkers are owned by Nordic Bioscience as well but are available from the corresponding author on reasonable request.
The study was conducted according to the Declaration of Helsinki and Good Clinical Practice guidelines; patients provided their informed consent. All local ethical committees approved the study, and written consent was obtained from all participants. Please see Additional file 1 for details about each ethical committee.
Not applicable.

Competing interests

JMBS, DJL, IB, ARB, and MAK are employees and MAK and ARB are shareholders of Nordic Bioscience. RT-S and BEM are employees and shareholders of GSK. PL reports grants and personal fees from Almirall, Astra Zeneca, Boehringer-Ingelheim, Novartis, GlaxoSmithKline, and personal fees from Norpharma, Takeda, Pfizer, and TEVA, outside the submitted work. JV has received honoraria for presenting and advising from Astra Zeneca, Boehringer-Ingelheim, Chiesi, GlaxoSmithKline and Novartis, outside the submitted work. The study was sponsored by GlaxoSmithKline; the Danish Agency for Science, Technology and Innovation; and the Danish Research Foundation.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Hogg JC, McDonough JE, Gosselink JV, Hayashi S. What drives the peripheral lung-remodeling process in chronic obstructive pulmonary disease? Proc Am Thorac Soc. 2009;6:668–72.CrossRefPubMedPubMedCentral Hogg JC, McDonough JE, Gosselink JV, Hayashi S. What drives the peripheral lung-remodeling process in chronic obstructive pulmonary disease? Proc Am Thorac Soc. 2009;6:668–72.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Burney P, Jithoo A, Kato B, Janson C, Mannino D, Nizankowska-Mogilnicka E, et al. Chronic obstructive pulmonary disease mortality and prevalence: the associations with smoking and poverty--a BOLD analysis. Thorax. 2014;69:465–73.CrossRefPubMed Burney P, Jithoo A, Kato B, Janson C, Mannino D, Nizankowska-Mogilnicka E, et al. Chronic obstructive pulmonary disease mortality and prevalence: the associations with smoking and poverty--a BOLD analysis. Thorax. 2014;69:465–73.CrossRefPubMed
3.
Zurück zum Zitat Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347–65.CrossRefPubMed Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary. Am J Respir Crit Care Med. 2013;187:347–65.CrossRefPubMed
4.
Zurück zum Zitat Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18:381–8.CrossRefPubMed Segreti A, Stirpe E, Rogliani P, Cazzola M. Defining phenotypes in COPD: an aid to personalized healthcare. Mol Diagn Ther. 2014;18:381–8.CrossRefPubMed
5.
Zurück zum Zitat Calverley PM. New treatments for COPD: many miles still to go. Lancet Respir Med. 2014;2:6–7.CrossRefPubMed Calverley PM. New treatments for COPD: many miles still to go. Lancet Respir Med. 2014;2:6–7.CrossRefPubMed
6.
Zurück zum Zitat Karsdal MA, Krarup H, Sand JM, Christensen PB, Gerstoft J, Leeming DJ, et al. Review article: the efficacy of biomarkers in chronic fibroproliferative diseases - early diagnosis and prognosis, with liver fibrosis as an exemplar. Aliment Pharmacol Ther. 2014;40:233–49.CrossRefPubMed Karsdal MA, Krarup H, Sand JM, Christensen PB, Gerstoft J, Leeming DJ, et al. Review article: the efficacy of biomarkers in chronic fibroproliferative diseases - early diagnosis and prognosis, with liver fibrosis as an exemplar. Aliment Pharmacol Ther. 2014;40:233–49.CrossRefPubMed
7.
Zurück zum Zitat Salazar LM, Herrera AM. Fibrotic response of tissue remodeling in COPD. Lung. 2011;189:101–9.CrossRefPubMed Salazar LM, Herrera AM. Fibrotic response of tissue remodeling in COPD. Lung. 2011;189:101–9.CrossRefPubMed
8.
Zurück zum Zitat Karsdal MA, Nielsen MJ, Sand JM, Henriksen K, Genovese F, Bay-Jensen AC, et al. Extracellular matrix remodeling: the common denominator in connective tissue diseases. Possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev Technol. 2013;11:70–92.CrossRefPubMedPubMedCentral Karsdal MA, Nielsen MJ, Sand JM, Henriksen K, Genovese F, Bay-Jensen AC, et al. Extracellular matrix remodeling: the common denominator in connective tissue diseases. Possibilities for evaluation and current understanding of the matrix as more than a passive architecture, but a key player in tissue failure. Assay Drug Dev Technol. 2013;11:70–92.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Kranenburg AR, Willems-Widyastuti A, Moori WJ, Sterk PJ, Alagappan VK, de Boer WI, et al. Enhanced bronchial expression of extracellular matrix proteins in chronic obstructive pulmonary disease. Am J Clin Pathol. 2006;126:725–35.CrossRefPubMed Kranenburg AR, Willems-Widyastuti A, Moori WJ, Sterk PJ, Alagappan VK, de Boer WI, et al. Enhanced bronchial expression of extracellular matrix proteins in chronic obstructive pulmonary disease. Am J Clin Pathol. 2006;126:725–35.CrossRefPubMed
11.
Zurück zum Zitat Finlay GA, O'Driscoll LR, Russell KJ, D'Arcy EM, Masterson JB, Fitzgerald MX, et al. Matrix metalloproteinase expression and production by alveolar macrophages in emphysema. Am J Respir Crit Care Med. 1997;156:240–7.CrossRefPubMed Finlay GA, O'Driscoll LR, Russell KJ, D'Arcy EM, Masterson JB, Fitzgerald MX, et al. Matrix metalloproteinase expression and production by alveolar macrophages in emphysema. Am J Respir Crit Care Med. 1997;156:240–7.CrossRefPubMed
12.
Zurück zum Zitat Hallgren O, Nihlberg K, Dahlback M, Bjermer L, Eriksson LT, Erjefalt JS, et al. Altered fibroblast proteoglycan production in COPD. Respir Res. 2010;11:55.CrossRefPubMedPubMedCentral Hallgren O, Nihlberg K, Dahlback M, Bjermer L, Eriksson LT, Erjefalt JS, et al. Altered fibroblast proteoglycan production in COPD. Respir Res. 2010;11:55.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Sandhaus RA, Turino G. Neutrophil elastase-mediated lung disease. COPD. 2013;10(Suppl 1):60–3.CrossRefPubMed Sandhaus RA, Turino G. Neutrophil elastase-mediated lung disease. COPD. 2013;10(Suppl 1):60–3.CrossRefPubMed
14.
Zurück zum Zitat Churg A, Zhou S, Wright JL. Series "matrix metalloproteinases in lung health and disease": matrix metalloproteinases in COPD. Eur Respir J. 2012;39:197–209.CrossRefPubMed Churg A, Zhou S, Wright JL. Series "matrix metalloproteinases in lung health and disease": matrix metalloproteinases in COPD. Eur Respir J. 2012;39:197–209.CrossRefPubMed
15.
Zurück zum Zitat Cane JL, Mallia-Millanes B, Forrester DL, Knox AJ, Bolton CE, Johnson SR. Matrix metalloproteinases −8 and −9 in the airways, blood and urine during exacerbations of COPD. COPD. 2016;13:26–34.CrossRefPubMed Cane JL, Mallia-Millanes B, Forrester DL, Knox AJ, Bolton CE, Johnson SR. Matrix metalloproteinases −8 and −9 in the airways, blood and urine during exacerbations of COPD. COPD. 2016;13:26–34.CrossRefPubMed
16.
Zurück zum Zitat van Kuppevelt TH, Veerkamp JH, Timmermans JA. Immunoquantification of type I, III, IV and V collagen in small samples of human lung parenchyma. Int J Biochem Cell Biol. 1995;27:775–82.CrossRefPubMed van Kuppevelt TH, Veerkamp JH, Timmermans JA. Immunoquantification of type I, III, IV and V collagen in small samples of human lung parenchyma. Int J Biochem Cell Biol. 1995;27:775–82.CrossRefPubMed
17.
Zurück zum Zitat Suki B, Ito S, Stamenovic D, Lutchen KR, Ingenito EP. Biomechanics of the lung parenchyma: critical roles of collagen and mechanical forces. J Appl Physiol. 2005;98:1892–9.CrossRefPubMed Suki B, Ito S, Stamenovic D, Lutchen KR, Ingenito EP. Biomechanics of the lung parenchyma: critical roles of collagen and mechanical forces. J Appl Physiol. 2005;98:1892–9.CrossRefPubMed
19.
Zurück zum Zitat Veidal SS, Karsdal MA, Vassiliadis E, Nawrocki A, Larsen MR, Nguyen QH, et al. MMP mediated degradation of type VI collagen is highly associated with liver fibrosis--identification and validation of a novel biochemical marker assay. PLoS One. 2011;6:e24753.CrossRefPubMedPubMedCentral Veidal SS, Karsdal MA, Vassiliadis E, Nawrocki A, Larsen MR, Nguyen QH, et al. MMP mediated degradation of type VI collagen is highly associated with liver fibrosis--identification and validation of a novel biochemical marker assay. PLoS One. 2011;6:e24753.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Abdillahi SM, Bober M, Nordin S, Hallgren O, Baumgarten M, Erjefalt J, et al. Collagen VI is Upregulated in COPD and serves both as an adhesive target and a bactericidal barrier for Moraxella catarrhalis. J Innate Immun. 2015;7:506–17.CrossRefPubMed Abdillahi SM, Bober M, Nordin S, Hallgren O, Baumgarten M, Erjefalt J, et al. Collagen VI is Upregulated in COPD and serves both as an adhesive target and a bactericidal barrier for Moraxella catarrhalis. J Innate Immun. 2015;7:506–17.CrossRefPubMed
21.
Zurück zum Zitat Jenkins RG, Simpson JK, Saini G, Bentley JH, Russell AM, Braybrooke R, et al. Longitudinal change in collagen degradation biomarkers in idiopathic pulmonary fibrosis: an analysis from the prospective, multicentre PROFILE study. Lancet Respir Med. 2015;3:462–72.CrossRefPubMed Jenkins RG, Simpson JK, Saini G, Bentley JH, Russell AM, Braybrooke R, et al. Longitudinal change in collagen degradation biomarkers in idiopathic pulmonary fibrosis: an analysis from the prospective, multicentre PROFILE study. Lancet Respir Med. 2015;3:462–72.CrossRefPubMed
22.
Zurück zum Zitat Leeming DJ, Delling G, Koizumi M, Henriksen K, Karsdal MA, Li B, et al. Alpha CTX as a biomarker of skeletal invasion of breast cancer: immunolocalization and the load dependency of urinary excretion. Cancer Epidemiol Biomark Prev. 2006;15:1392–5.CrossRef Leeming DJ, Delling G, Koizumi M, Henriksen K, Karsdal MA, Li B, et al. Alpha CTX as a biomarker of skeletal invasion of breast cancer: immunolocalization and the load dependency of urinary excretion. Cancer Epidemiol Biomark Prev. 2006;15:1392–5.CrossRef
23.
Zurück zum Zitat Leeming DJ, Byrjalsen I, Jimenez W, Christiansen C, Karsdal MA. Protein fingerprinting of the extracellular matrix remodelling in a rat model of liver fibrosis--a serological evaluation. Liver Int. 2013;33:439–47.CrossRefPubMed Leeming DJ, Byrjalsen I, Jimenez W, Christiansen C, Karsdal MA. Protein fingerprinting of the extracellular matrix remodelling in a rat model of liver fibrosis--a serological evaluation. Liver Int. 2013;33:439–47.CrossRefPubMed
24.
Zurück zum Zitat Brasso K, Christensen IJ, Johansen JS, Teisner B, Garnero P, Price PA, et al. Prognostic value of PINP, bone alkaline phosphatase, CTX-I, and YKL-40 in patients with metastatic prostate carcinoma. Prostate. 2006;66:503–13.CrossRefPubMed Brasso K, Christensen IJ, Johansen JS, Teisner B, Garnero P, Price PA, et al. Prognostic value of PINP, bone alkaline phosphatase, CTX-I, and YKL-40 in patients with metastatic prostate carcinoma. Prostate. 2006;66:503–13.CrossRefPubMed
25.
Zurück zum Zitat Peris P, Alvarez L, Vidal S, Kasper D, Leeming DJ, Monegal A, et al. Biochemical response to bisphosphonate therapy in pagetic patients with skull involvement. Calcif Tissue Int. 2006;79:22–6.CrossRefPubMed Peris P, Alvarez L, Vidal S, Kasper D, Leeming DJ, Monegal A, et al. Biochemical response to bisphosphonate therapy in pagetic patients with skull involvement. Calcif Tissue Int. 2006;79:22–6.CrossRefPubMed
26.
Zurück zum Zitat Garnero P. Biomarkers for osteoporosis management: utility in diagnosis, fracture risk prediction and therapy monitoring. Mol Diagn Ther. 2008;12:157–70.CrossRefPubMed Garnero P. Biomarkers for osteoporosis management: utility in diagnosis, fracture risk prediction and therapy monitoring. Mol Diagn Ther. 2008;12:157–70.CrossRefPubMed
27.
Zurück zum Zitat Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, et al. Enzyme-linked immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen neoepitope, CIIM-increased serum CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem. 2011;44:423–9.CrossRefPubMed Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, et al. Enzyme-linked immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen neoepitope, CIIM-increased serum CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem. 2011;44:423–9.CrossRefPubMed
28.
Zurück zum Zitat Nielsen MJ, Nedergaard AF, Sun S, Veidal SS, Larsen L, Zheng Q, et al. The neo-epitope specific PRO-C3 ELISA measures true formation of type III collagen associated with liver and muscle parameters. Am J Transl Res. 2013;5:303–15.PubMedPubMedCentral Nielsen MJ, Nedergaard AF, Sun S, Veidal SS, Larsen L, Zheng Q, et al. The neo-epitope specific PRO-C3 ELISA measures true formation of type III collagen associated with liver and muscle parameters. Am J Transl Res. 2013;5:303–15.PubMedPubMedCentral
29.
Zurück zum Zitat Nielsen MJ, Kazankov K, Leeming DJ, Karsdal MA, Krag A, Barrera F, et al. Markers of collagen remodeling detect clinically significant fibrosis in chronic hepatitis C patients. PLoS One. 2015;10:e0137302.CrossRefPubMedPubMedCentral Nielsen MJ, Kazankov K, Leeming DJ, Karsdal MA, Krag A, Barrera F, et al. Markers of collagen remodeling detect clinically significant fibrosis in chronic hepatitis C patients. PLoS One. 2015;10:e0137302.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, Leeming DJ, Dam EB, Zheng Q, Qvist P, Karsdal MA. Enzyme-linked immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen neoepitope, CIIM--increased serum CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem. 2011;44(5-6):423–9. doi:10.1016/j.clinbiochem.2011.01.001. Epub 2011 Jan 9. PubMed PMID: 21223960. Bay-Jensen AC, Liu Q, Byrjalsen I, Li Y, Wang J, Pedersen C, Leeming DJ, Dam EB, Zheng Q, Qvist P, Karsdal MA. Enzyme-linked immunosorbent assay (ELISAs) for metalloproteinase derived type II collagen neoepitope, CIIM--increased serum CIIM in subjects with severe radiographic osteoarthritis. Clin Biochem. 2011;44(5-6):423–9. doi:10.​1016/​j.​clinbiochem.​2011.​01.​001. Epub 2011 Jan 9. PubMed PMID: 21223960.
31.
Zurück zum Zitat Bay-Jensen AC, Byrjalsen I, Siebuhr AS, Christiansen C, Platt A, Karsdal MA. Serological biomarkers of joint tissue turnover predict tocilizumab response at baseline. J Clin Rheumatol. 2014;20:332–5.PubMedPubMedCentral Bay-Jensen AC, Byrjalsen I, Siebuhr AS, Christiansen C, Platt A, Karsdal MA. Serological biomarkers of joint tissue turnover predict tocilizumab response at baseline. J Clin Rheumatol. 2014;20:332–5.PubMedPubMedCentral
32.
Zurück zum Zitat Sand JM, Knox AJ, Lange P, Sun S, Kristensen JH, Leeming DJ, et al. Accelerated extracellular matrix turnover during exacerbations of COPD. Respir Res. 2015;16:69.CrossRefPubMedPubMedCentral Sand JM, Knox AJ, Lange P, Sun S, Kristensen JH, Leeming DJ, et al. Accelerated extracellular matrix turnover during exacerbations of COPD. Respir Res. 2015;16:69.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Stolz D, Leeming DJ, Kristensen JH, Karsdal MA, Boersma W, Louis R, Milenkovic B, Kostikas K, Blasi F, Aerts J, Sand JM, Wouters EF, Rohde G, Prat C, Torres A, Welte T, Roth M, Papakonstantinou E, Tamm M. Systemic Biomarkers of Collagen and Elastin Turnover Are Associated With Clinically Relevant Outcomes in COPD. Chest. 2017;151(1):47–59. doi:10.1016/j.chest.2016.08.1440. Epub 2016 Aug 26. PubMed PMID: 27575358. Stolz D, Leeming DJ, Kristensen JH, Karsdal MA, Boersma W, Louis R, Milenkovic B, Kostikas K, Blasi F, Aerts J, Sand JM, Wouters EF, Rohde G, Prat C, Torres A, Welte T, Roth M, Papakonstantinou E, Tamm M. Systemic Biomarkers of Collagen and Elastin Turnover Are Associated With Clinically Relevant Outcomes in COPD. Chest. 2017;151(1):47–59. doi:10.​1016/​j.​chest.​2016.​08.​1440. Epub 2016 Aug 26. PubMed PMID: 27575358.
34.
Zurück zum Zitat Sand JM, Martinez G, Midjord AK, Karsdal MA, Leeming DJ, Lange P. Characterization of serological neo-epitope biomarkers reflecting collagen remodeling in clinically stable chronic obstructive pulmonary disease. Clin Biochem. 2016;49:1144–51.CrossRefPubMed Sand JM, Martinez G, Midjord AK, Karsdal MA, Leeming DJ, Lange P. Characterization of serological neo-epitope biomarkers reflecting collagen remodeling in clinically stable chronic obstructive pulmonary disease. Clin Biochem. 2016;49:1144–51.CrossRefPubMed
35.
Zurück zum Zitat Sand JM, Leeming DJ, Byrjalsen I, Bihlet AR, Lange P, Tal-Singer R, et al. High levels of biomarkers of collagen remodeling are associated with increased mortality in C. Respir Res. 2016;17:125.CrossRefPubMedPubMedCentral Sand JM, Leeming DJ, Byrjalsen I, Bihlet AR, Lange P, Tal-Singer R, et al. High levels of biomarkers of collagen remodeling are associated with increased mortality in C. Respir Res. 2016;17:125.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Vestbo J, Edwards LD, Scanlon PD, Yates JC, Agusti A, Bakke P, et al. Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med. 2011;365:1184–92.CrossRefPubMed Vestbo J, Edwards LD, Scanlon PD, Yates JC, Agusti A, Bakke P, et al. Changes in forced expiratory volume in 1 second over time in COPD. N Engl J Med. 2011;365:1184–92.CrossRefPubMed
37.
Zurück zum Zitat Leeming D, He Y, Veidal S, Nguyen Q, Larsen D, Koizumi M, et al. A novel marker for assessment of liver matrix remodeling: an enzyme-linked immunosorbent assay (ELISA) detecting a MMP generated type I collagen neo-epitope (C1M). Biomarkers. 2011;16:616–28.CrossRefPubMed Leeming D, He Y, Veidal S, Nguyen Q, Larsen D, Koizumi M, et al. A novel marker for assessment of liver matrix remodeling: an enzyme-linked immunosorbent assay (ELISA) detecting a MMP generated type I collagen neo-epitope (C1M). Biomarkers. 2011;16:616–28.CrossRefPubMed
38.
Zurück zum Zitat Vestbo J, Anderson W, Coxson HO, Crim C, Dawber F, Edwards L, et al. Evaluation of COPD longitudinally to identify predictive surrogate end-points (ECLIPSE). Eur Respir J. 2008;31:869–73.CrossRefPubMed Vestbo J, Anderson W, Coxson HO, Crim C, Dawber F, Edwards L, et al. Evaluation of COPD longitudinally to identify predictive surrogate end-points (ECLIPSE). Eur Respir J. 2008;31:869–73.CrossRefPubMed
39.
Zurück zum Zitat Celli BR, Locantore N, Yates J, Tal-Singer R, Miller BE, Bakke P, et al. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185:1065–72.CrossRefPubMed Celli BR, Locantore N, Yates J, Tal-Singer R, Miller BE, Bakke P, et al. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2012;185:1065–72.CrossRefPubMed
40.
Zurück zum Zitat Decramer M, Celli B, Kesten S, Lystig T, Mehra S, Tashkin DP. Effect of tiotropium on outcomes in patients with moderate chronic obstructive pulmonary disease (UPLIFT): a prespecified subgroup analysis of a randomised controlled trial. Lancet. 2009;374:1171–8.CrossRefPubMed Decramer M, Celli B, Kesten S, Lystig T, Mehra S, Tashkin DP. Effect of tiotropium on outcomes in patients with moderate chronic obstructive pulmonary disease (UPLIFT): a prespecified subgroup analysis of a randomised controlled trial. Lancet. 2009;374:1171–8.CrossRefPubMed
41.
Zurück zum Zitat Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775–89.CrossRefPubMed Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356:775–89.CrossRefPubMed
42.
Zurück zum Zitat Celli BR, Thomas NE, Anderson JA, Ferguson GT, Jenkins CR, Jones PW, et al. Effect of pharmacotherapy on rate of decline of lung function in chronic obstructive pulmonary disease: results from the TORCH study. Am J Respir Crit Care Med. 2008;178:332–8.CrossRefPubMed Celli BR, Thomas NE, Anderson JA, Ferguson GT, Jenkins CR, Jones PW, et al. Effect of pharmacotherapy on rate of decline of lung function in chronic obstructive pulmonary disease: results from the TORCH study. Am J Respir Crit Care Med. 2008;178:332–8.CrossRefPubMed
43.
Zurück zum Zitat Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543–54.CrossRefPubMed Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008;359:1543–54.CrossRefPubMed
44.
Zurück zum Zitat Lange P, Celli B, Agusti A. Lung-function trajectories and chronic obstructive pulmonary disease. N Engl J Med. 2015;373:1575.CrossRefPubMed Lange P, Celli B, Agusti A. Lung-function trajectories and chronic obstructive pulmonary disease. N Engl J Med. 2015;373:1575.CrossRefPubMed
45.
Zurück zum Zitat Bihlet AR, Karsdal MA, Bay-Jensen AC, Read S, Kristensen JH, Sand JM, et al. Clinical drug development using dynamic biomarkers to enable personalized health care in COPD. Chest. 2015;148:16–23.CrossRefPubMed Bihlet AR, Karsdal MA, Bay-Jensen AC, Read S, Kristensen JH, Sand JM, et al. Clinical drug development using dynamic biomarkers to enable personalized health care in COPD. Chest. 2015;148:16–23.CrossRefPubMed
46.
Zurück zum Zitat Vestbo J, Rennard S. Chronic obstructive pulmonary disease biomarker(s) for disease activity needed--urgently. Am J Respir Crit Care Med. 2010;182:863–4.CrossRefPubMed Vestbo J, Rennard S. Chronic obstructive pulmonary disease biomarker(s) for disease activity needed--urgently. Am J Respir Crit Care Med. 2010;182:863–4.CrossRefPubMed
47.
Zurück zum Zitat Faner R, Tal-Singer R, Riley JH, Celli B, Vestbo J, MacNee W, et al. Lessons from ECLIPSE: a review of COPD biomarkers. Thorax. 2014;69:666–72.CrossRefPubMed Faner R, Tal-Singer R, Riley JH, Celli B, Vestbo J, MacNee W, et al. Lessons from ECLIPSE: a review of COPD biomarkers. Thorax. 2014;69:666–72.CrossRefPubMed
48.
Zurück zum Zitat Luisetti M, Ma S, Iadarola P, Stone PJ, Viglio S, Casado B, et al. Desmosine as a biomarker of elastin degradation in COPD: current status and future directions. Eur Respir J. 2008;32:1146–57.CrossRefPubMed Luisetti M, Ma S, Iadarola P, Stone PJ, Viglio S, Casado B, et al. Desmosine as a biomarker of elastin degradation in COPD: current status and future directions. Eur Respir J. 2008;32:1146–57.CrossRefPubMed
49.
Zurück zum Zitat Yokoyama A, Kohno N, Hamada H, Sakatani M, Ueda E, Kondo K, et al. Circulating KL-6 predicts the outcome of rapidly progressive idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 1998;158:1680–4.CrossRefPubMed Yokoyama A, Kohno N, Hamada H, Sakatani M, Ueda E, Kondo K, et al. Circulating KL-6 predicts the outcome of rapidly progressive idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 1998;158:1680–4.CrossRefPubMed
50.
Zurück zum Zitat Prasse A, Pechkovsky DV, Toews GB, Schafer M, Eggeling S, Ludwig C, et al. CCL18 as an indicator of pulmonary fibrotic activity in idiopathic interstitial pneumonias and systemic sclerosis. Arthritis Rheum. 2007;56:1685–93.CrossRefPubMed Prasse A, Pechkovsky DV, Toews GB, Schafer M, Eggeling S, Ludwig C, et al. CCL18 as an indicator of pulmonary fibrotic activity in idiopathic interstitial pneumonias and systemic sclerosis. Arthritis Rheum. 2007;56:1685–93.CrossRefPubMed
51.
Zurück zum Zitat Greene KE, King TE Jr, Kuroki Y, Bucher-Bartelson B, Hunninghake GW, Newman LS, et al. Serum surfactant proteins-a and -D as biomarkers in idiopathic pulmonary fibrosis. Eur Respir J. 2002;19:439–46.CrossRefPubMed Greene KE, King TE Jr, Kuroki Y, Bucher-Bartelson B, Hunninghake GW, Newman LS, et al. Serum surfactant proteins-a and -D as biomarkers in idiopathic pulmonary fibrosis. Eur Respir J. 2002;19:439–46.CrossRefPubMed
52.
Zurück zum Zitat Rabinovich RA, Miller BE, Wrobel K, Ranjit K, Williams MC, Drost E, et al. Circulating desmosine levels do not predict emphysema progression but are associated with cardiovascular risk and mortality in COPD. Eur Respir J. 2016;47:1365–73.CrossRefPubMed Rabinovich RA, Miller BE, Wrobel K, Ranjit K, Williams MC, Drost E, et al. Circulating desmosine levels do not predict emphysema progression but are associated with cardiovascular risk and mortality in COPD. Eur Respir J. 2016;47:1365–73.CrossRefPubMed
53.
Zurück zum Zitat Mannino DM, Tal-Singer R, Lomas DA, Vestbo J, Graham BR, Tetzlaff K, et al. Plasma fibrinogen as a biomarker for mortality and hospitalized exacerbations in people with COPD. Chronic Obstr Pulm Dis (Miami ). 2015;2:23–34.CrossRef Mannino DM, Tal-Singer R, Lomas DA, Vestbo J, Graham BR, Tetzlaff K, et al. Plasma fibrinogen as a biomarker for mortality and hospitalized exacerbations in people with COPD. Chronic Obstr Pulm Dis (Miami ). 2015;2:23–34.CrossRef
54.
Zurück zum Zitat Leeming DJ, Sand JM, Nielsen MJ, Genovese F, Martinez FJ, Hogaboam CM, et al. Serological investigation of the collagen degradation profile of patients with chronic obstructive pulmonary disease or idiopathic pulmonary fibrosis. Biomark Insights. 2012;7:119–26.CrossRefPubMedPubMedCentral Leeming DJ, Sand JM, Nielsen MJ, Genovese F, Martinez FJ, Hogaboam CM, et al. Serological investigation of the collagen degradation profile of patients with chronic obstructive pulmonary disease or idiopathic pulmonary fibrosis. Biomark Insights. 2012;7:119–26.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Sand JM, Leeming DJ, Byrjalsen I, Bihlet AR, Lange P, Tal-Singer R, Miller BE, Karsdal MA, Vestbo J. High levels of biomarkers of collagen remodeling are associated with increased mortality in COPD - results from the ECLIPSE study. Respir Res. 2016;17(1):125. PubMed PMID: 27716343; PubMed Central PMCID: PMC5050854. Sand JM, Leeming DJ, Byrjalsen I, Bihlet AR, Lange P, Tal-Singer R, Miller BE, Karsdal MA, Vestbo J. High levels of biomarkers of collagen remodeling are associated with increased mortality in COPD - results from the ECLIPSE study. Respir Res. 2016;17(1):125. PubMed PMID: 27716343; PubMed Central PMCID: PMC5050854.
56.
Zurück zum Zitat Vestbo J, Agusti A, Wouters EF, Bakke P, Calverley PM, Celli B, 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;189:1022–30.CrossRefPubMed Vestbo J, Agusti A, Wouters EF, Bakke P, Calverley PM, Celli B, 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;189:1022–30.CrossRefPubMed
57.
Zurück zum Zitat Deng ZC, Zhao P, Cao C, Sun SF, Zhao F, Lu CY, et al. C-reactive protein as a prognostic marker in chronic obstructive pulmonary disease. Exp Ther Med. 2014;7:443–6.CrossRefPubMed Deng ZC, Zhao P, Cao C, Sun SF, Zhao F, Lu CY, et al. C-reactive protein as a prognostic marker in chronic obstructive pulmonary disease. Exp Ther Med. 2014;7:443–6.CrossRefPubMed
58.
Zurück zum Zitat 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;175:250–5.CrossRefPubMed 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;175:250–5.CrossRefPubMed
59.
Zurück zum Zitat Agusti A, Edwards LD, Rennard SI, MacNee W, Tal-Singer R, Miller BE, et al. Persistent systemic inflammation is associated with poor clinical outcomes in COPD: a novel phenotype. PLoS One. 2012;7:e37483.CrossRefPubMedPubMedCentral Agusti A, Edwards LD, Rennard SI, MacNee W, Tal-Singer R, Miller BE, et al. Persistent systemic inflammation is associated with poor clinical outcomes in COPD: a novel phenotype. PLoS One. 2012;7:e37483.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Lange P, Celli B, Agusti A, Boje JG, Divo M, Faner R, et al. Lung-function trajectories leading to chronic obstructive pulmonary disease. N Engl J Med. 2015;373:111–22.CrossRefPubMed Lange P, Celli B, Agusti A, Boje JG, Divo M, Faner R, et al. Lung-function trajectories leading to chronic obstructive pulmonary disease. N Engl J Med. 2015;373:111–22.CrossRefPubMed
61.
Zurück zum Zitat Ferrari R, Tanni SE, Caram LM, Correa C, Correa CR, Godoy I. Three-year follow-up of interleukin 6 and C-reactive protein in chronic obstructive pulmonary disease. Respir Res. 2013;14:24.CrossRefPubMedPubMedCentral Ferrari R, Tanni SE, Caram LM, Correa C, Correa CR, Godoy I. Three-year follow-up of interleukin 6 and C-reactive protein in chronic obstructive pulmonary disease. Respir Res. 2013;14:24.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Shaaban R, Kony S, Driss F, Leynaert B, Soussan D, Pin I, et al. Change in C-reactive protein levels and FEV1 decline: a longitudinal population-based study. Respir Med. 2006;100:2112–20.CrossRefPubMed Shaaban R, Kony S, Driss F, Leynaert B, Soussan D, Pin I, et al. Change in C-reactive protein levels and FEV1 decline: a longitudinal population-based study. Respir Med. 2006;100:2112–20.CrossRefPubMed
63.
Zurück zum Zitat Karsdal MA, Henriksen K, Leeming DJ, Woodworth T, Vassiliadis E, Bay-Jensen AC. Novel combinations of post-translational modification (PTM) neo-epitopes provide tissue-specific biochemical markers--are they the cause or the consequence of the disease? Clin Biochem. 2010;43:793–804.CrossRefPubMed Karsdal MA, Henriksen K, Leeming DJ, Woodworth T, Vassiliadis E, Bay-Jensen AC. Novel combinations of post-translational modification (PTM) neo-epitopes provide tissue-specific biochemical markers--are they the cause or the consequence of the disease? Clin Biochem. 2010;43:793–804.CrossRefPubMed
Metadaten
Titel
Biomarkers of collagen turnover are related to annual change in FEV1 in patients with chronic obstructive pulmonary disease within the ECLIPSE study
verfasst von
Diana J. Leeming
Inger Byrjalsen
Jannie M. B. Sand
Asger R. Bihlet
Peter Lange
Ruth Thal-Singer
Bruce E. Miller
Morten A. Karsdal
Jørgen Vestbo
The Evaluation of COPD Longitudinally to Identify Surrogate Endpoints (ECLIPSE) study investigators
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2017
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-017-0505-4

Weitere Artikel der Ausgabe 1/2017

BMC Pulmonary Medicine 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.