Skip to main content
Erschienen in: Respiratory Research 1/2017

Open Access 01.12.2017 | Review

Neutrophil elastase in bronchiectasis

verfasst von: Andrea Gramegna, Francesco Amati, Leonardo Terranova, Giovanni Sotgiu, Paolo Tarsia, Daniela Miglietta, Maria Adelaide Calderazzo, Stefano Aliberti, Francesco Blasi

Erschienen in: Respiratory Research | Ausgabe 1/2017

Abstract

The role of neutrophil elastase (NE) is poorly understood in bronchiectasis because of the lack of preclinical data and so most of the assumptions made about NE inhibitor potential benefit is based on data from CF. In this context, NE seems to be a predictor of long-term clinical outcomes and a possible target of treatment. In order to better evaluate the role of NE in bronchiectasis, a systematic search of scientific evidence was performed.
Two investigators independently performed the search on PubMed and included studies published up to May 15, 2017 according to predefined criteria. A final pool of 31 studies was included in the systematic review, with a total of 2679 patients. For each paper data of interest were extracted and reported in table.
In this review sputum NE has proved useful as an inflammatory marker both in stable state bronchiectasis and during exacerbations and local or systemic antibiotic treatment. NE has also been associated with risk of exacerbation, time to next exacerbation and all-cause mortality. This study reviews also the role of NE as a specific target of treatment in bronchiectasis. Inhibition of NE is at a very early stage and future interventional studies should evaluate safety and efficacy for new molecules and formulations.

Background

Neutrophil elastase

Neutrophils play a key role in the pathogenesis of numerous diseases, ranging from chronic inflammatory disorders to infectious diseases and neoplasms [1]. Their role in the innate immune system mainly consists in extracellular phagocytosis, involving reactive oxygen species, cationic proteins, and enzymes. Several granules are sequentially formed in neutrophil cytoplasm, differing in their propensity for exocytosis, according to their biosynthesis timing. Granules formed at a later stage of differentiation shows a higher secretory potential than those formed in immature myeloid cells, such as the azurophilic granules containing myeloperoxidase, defensins, and proteinases. Neutrophils live shortly in the blood of healthy individuals (around 8 h), but following inflammatory conditions they become activated and their longevity may increase up to 5 days. The prolonged life of neutrophils leads to functional changes and contributes to the inflammatory-associated morbidity shown in bronchiectasis patients [2].
Once inflammatory trigger develops in peripheral tissues, neutrophils are rapidly recruited toward to the anatomical site of inflammation. Their migration out of the blood stream involves endothelial attachment and rolling along the endothelium, activation, adhesion, and, finally, trans-endothelial extravasation into the interstitium. The neutrophilic inflammatory response depends on the degranulation of pre-formed mediators from cytoplasmatic granules and on the ability to generate respiratory burst activity via reactive oxygen species system. Although they ontogenetically play key functions in antimicrobial defense, their activation can also damage host cells and tissues. Neutrophil degranulation is responsible for the release of several inflammatory mediators, such as neutrophil elastase, cathepsin G, and proteinase 3 [3].
Human neutrophil elastase (NE) is a proteolytic enzyme belonging to the chymotrypsin-like family of the serine-proteinases. NE is a 218 aminoacid long protein packaged in cytoplasmatic azurophilic granules in neutrophil granulocytes [4]. Though NE is mainly involved in the response against bacteria, it can cause several detrimental effects, including extracellular matrix destruction, mucus gland hyperplasia and increased mucus production, reduction of ciliary beating rate, and direct damage to the airway epithelium [57]. The acid milieu within granules protects the cell from proteolytic activity. Upon neutrophil activation, degranulation is triggered by the mild alkalosis resulting from the fusion between the granules and cytoplasmic phagosomes containing engulfed bacteria [8]. In addition to this intracellular pathway, NE is also liberated into the extracellular space, both free and membrane-bound. High concentrations of soluble NE participate in bacterial killing at the site of degranulation and in a small area around the cell. More distantly, activated enzymes are completely neutralized by proteinase inhibitors. On the other hand, membrane-bound NE is remarkably resistant to inhibition by the anti-proteinase system, thus preserving its proteolytic activity. Overexpression of this mechanism has the potential to cause tissue damage [9].
During neutrophil activation, NE-containing granules also translocate from the cytoplasm to the nucleus, where it is involved in the degradation of chromatin by splitting histones. Then, neutrophils disintegrate and release neutrophil extracellular traps (NETs). Although this remains a controversial or fully unexplored area, this process is hypothesized to facilitate the neutralization of pathogens, including bacteria, fungi, and protozoa. NETs are networks of extracellular fibers, composed of DNA bound to histone proteins and neutrophil proteinases. NE is the most common non-histone protein in NETs [10].
A complex interaction among proteases and anti-proteases is required for an effective respiratory immune system. Lower airways in patients with neutrophil elastase-mediated lung diseases are characterized by an excess of proteases leading to tissue destruction and lung function decline. A1AT is the most abundant anti-protease within the lung and the major inhibitor of NE. A1AT is encoded by the SERPINA1 gene located on the long arm of chromosome 14 and produced mainly by hepatocytes and, to a lesser extent, by alveolar macrophages. Imbalance between NE and A1AT may be due to either an A1AT deficiency, as documented in COPD and emphysema, or unopposed NE activity, as proved in CF and bronchiectasis [11]. In the latter context, NE shows the ability to combine with polyanions (such as DNA or glycosaminoglycans) and syndecan-1 becoming inaccessible and limiting its inactivation by A1AT [12].
Besides NE, the serine protease group also includes cathepsin G (Cat G) and proteinase 3 (PR3), which are also stored in high concentrations in the azurophil granules within the neutrophil cytoplasm [4]. Cat G is a 235-amino acid protein released during the neutrophil activation process. Intracellularly, Cat G is released within the phagosome to degrade engulfed pathogens; extracellularly, it contributes to the degradation of structural ECM components and other immune mediators [13]. PR3 is a 222-amino acid long serine protease encoded by the PRTN3 gene and expressed by activated neutrophils. Its biological function is similar to that of NE. In particular, PR3 is responsible for proteolytic degradation and activation of IL-8 in a truncated peptide with fostered chemoattractant effect towards neutrophils.

NE determination assays

Different techniques have been used for NE determination. Both ready to use commercial kits and in-home assays are useful for in vitro quantitative determination of NE. The three most used techniques are ELISA [14], fluorimetric [15] and spectrophotometric [16] methods. In Table 1 some example of NE determination assays is provided showing different samples, types of measurement and detection ranges.
Table 1
Neutrophil elastase determination assays
Name
Samples
Type of measurement
Assay
Unit of measure
N° of test
Detection range
Limit of detection
Human Elastase ELISA (Hycult Biotech)
Plasma, Cell culture supernatant
Active NE
ELISA
ng/mL
2 × 96
0.4–25 ng/mL
0.4 ng/mL
ProteaseTag® Active NE Immunoassay (ProAxsis Ltd)
BAL, Sputum sol, Serum free media
Active NE
ELISA
ng/mL
96
15.625–1000 ng/mL
7.2 ng/mL
Human PMN Elastase ELISA Kit (Abcam)
Cell culture supernatant, Serum, Plasma
PMN Elastase/alpha1-PI complex
ELISA
ng/mL
96
0.16–10 ng/ml
1.98 pg/mL
Neutrophil elastase Human ELISA Kit, CE (eBioscience)
Cell culture supernatant, Serum, Plasma
PMN Elastase/alpha1-PI complex
ELISA
ng/mL
96
0.16–10.0 ng/mL
1.98 pg/mL
Neutrophil Elastase Activity Assay Kit (Abcam)
Plasma, Whole Blood, Purified protein
Active NE
Fluorometric
ng
100
1–25 ng
1 ng
Dynatech MR 5000 (Dynatech Corporation)
Sputum sol
Active NE
Spectrophotometry
mcgM
96
Neutrophil Elastase Activity Assay Kit (Cayman Chemical)
Blood
Active NE
Fluorometric
mU/mL
2 × 96
0–10 mU/mL
0.156 mU/mL
Definitions: NE neutrophil elastase, alpha1-PI alpha-1 proteinase inhibitor; PMN polymorphonuclear leukocytes
NE can be detected by two different methods: the assessment of NE ability to proteolytically cleave a synthetic substrate (qualitative measurement); and the quantification of human NE concentrations in different biological media (quantitative measurement). Both the detection of the active enzyme and the measurement of the amount of enzyme bound to its inhibitor, alpha-1 antitrypsin (A1AT), are techniques currently used to measure NE in biological samples.
When ELISA assays are employed, the wells in a microtiter plate are coated with a specific antibody able to bind NE. ProteaseTag® is a quantitative method to dose NE activity using NE-Tag and the subsequent antibody step provides additional signal amplification with increased sensitivity with good results in a recent study conducted by Chalmers [17].
Fluorimetric and spectrophotometric assays are based on the ability of NE to proteolytically cleave a synthetic substrate causing the release of a fluorophore or a change in optical density, respectively, which can be quantified by fluorescence or absorbance microplate readers. Standard curves are obtained for both methods and concentration of samples can be determined from curve interpretation.
Commercial kits may also be used to quantify NE in plasma, serum, cell-culture supernatant, bronchoalveolar lavage (BAL), sputum, or whole blood. Among other methodologies, elastin agarose diffusion plate for measurement of elastolytic activity has been also reported in previous literature [18].
Elastin degradation as a consequence of NE activity results in the presence of elastin-derived peptides, including desmosine and isodesmosine in serum, plasma and urine. Both separation based on high performance liquid chromatography/mass spectrometry and radioimmunoassay are validated techniques for the detection of iso- and desmosine in body fluids [19, 20]. Finally, high-performance capillary electrophoresis has been applied to the study of proteases [21].

Neutrophil elastase in chronic respiratory diseases

Cystic fibrosis

An outburst of neutrophilic inflammation characterizes CF patients and it is associated with high concentrations of neutrophil proteases, including NE. NE has several implications in inflammation in CF, although a comprehensive summary of NE effects is not in the purpose if this review NE has an important role in the pathogenesis of lung inflammation in CF even in the absence of any infections. High levels of both NE and IL-8 have been demonstrated in BAL of infants with CF [22]. A recent study by Sly showed a correlation between NE activity in BAL from CF children and the early development of bronchiectasis [23].
NE is a potent upregulator of several inflammatory chemokines, such as IL-8 and MMP-9, leading to a self-perpetuating cycle of neutrophilic inflammation with several detrimental effects [24]. Firstly, NE impairs ciliary beating and promotes expression of respiratory tract mucins (MUC5AC and MUC5B), resulting in muco-ciliary clearance failure [7, 25]. Secondly, NE-dependent structural damage against elastin and other pulmonary components leads to the irreversible airway dilation and early bronchiectasis development. Thirdly, NE can degradate lactoferrin, an important anti-microbial glycoprotein as well as several molecules involved in pathogens’ opsonization [2629]. Furthermore, NE can cause impairment of lymphocytic function by degradation of T-cell surface receptors and interfere with the process of antigen presentation by dendritic cells [30, 31]. The sum of all these mechanisms leads to the paradox of an overstimulated immune system unable to effectively kill colonizing pathogens.
Finally, NE may directly affect the ion transport in CF cells acting as a potent activator of silent ENaC channels causing increased Na absorption and further airway surface dehydration and muco-ciliary dysfunction [32]. In a recent study, Le Gars and coworkers illustrated that high NE levels lead to CFTR loss of function via activation of intracellular calpains that are directly responsible for the proteolytic degradation of CFTR [33].
Treatments targeted against NE have been evaluated in CF patients. AZD9668, a reversible oral inhibitor of NE, when administered 60 mg twice daily orally for 4 weeks, showed no effect on sputum neutrophil counts, NE activity, lung function or clinical outcomes, but a consistent pattern of reduction in sputum inflammatory biomarkers and a significant decrease of free and total urine desmosine were reported in a double-blind randomized controlled trial (RCT) enrolling ~60 CF patients [34]. A phase-II clinical trial assessed the efficacy of inhaled A1AT, with significant reduction of NE activity and inflammatory biomarker expression without any effects on lung function [35]. The limited efficacy observed with both AZD9668 and A1AT in CF patients could be mainly ascribed to the fact that the studies were most likely too short and not powered to investigate disease-modifying effects.

Chronic obstructive pulmonary disease

In patients with COPD neutrophils in sputum correlate with severity of disease in terms of FEV1 decline and peripheral airway dysfunction identified by high-resolution computed tomography [36]. Several studies highlighted that cigarette smoke and lung pollutants activate macrophages and epithelial cells triggering the release of neutrophil chemotactic factors [37, 38]. In this context NE is a major determinant of tissue damage. A landmark study on COPD-related lung tissue by Damiano showed a positive correlation between the distribution of NE within alveolar spaces and the presence of emphysema [39]. Instillation of human NE has been shown to induce emphysema in the animal model, while Shapiro and coworkers demonstrated that mice deficient for NE expression were protected from the development of emphysematous changes [40, 41]. More recently, Paone have found that elevated levels of NE in COPD in both sputum and BAL highly correlated with severity of disease and lung function decline [42].
Treatment strategies targeting NE have been proposed also for patients with COPD. AZD9668 was shown to slow lung function decline and dampen the inflammatory burden [43]. However, two RCTs which recruited symptomatic COPD patients exposed to AZD9668 for 12 weeks failed to show any clinical efficacy nor beneficial effects on inflammatory biomarker expression [44, 45]. This lack of efficacy may be due to inappropriate study design in term of patients selection, relevant dose, duration of treatment and endpoints assessments. It has been recently demonstrated that in a subpopulation with alpha1-antitrypsin (A1AT) deficiency, intravenous administration of A1AT slowed the loss of lung parenchyma [46].

Asthma

Chronic inflammation in asthma is highly heterogeneous. Although in the past asthma was thought as a mere Th2-mediated inflammatory response to inhaled antigens, neutrophils and their products have recently been considered to play a role during both stable conditions and exacerbations [47, 48]. Simpson and colleagues showed that neutrophilic asthma is characterized by higher concentrations of active NE, generating the hypothesis that anti-proteases may be beneficial in the treatment of asthmatic patients [49]. At the current state, no studies on NE inhibitors in patients with asthma are currently available.

Neutrophil Elastase in Bronchiectasis

Bronchiectasis is defined as an abnormal and permanent dilation of the bronchi associated with daily cough, sputum production, and recurrent respiratory infections [50]. The prevalence of bronchiectasis has risen during the last decade both in hospital and community settings owing to population aging, as well as widespread use of chest CT scan and more awareness of the disease [50, 51]. The pathophysiology of bronchiectasis is not completely understood, particularly in view of the heterogeneity of the disease and the absence of an animal model [52]. A fundamental role is played by both impaired mucus clearance and chronic bacterial infections leading to a serious neutrophilic activation by the release of chemotactic mediators. A recent study by Dente and coworkers demonstrated that sputum neutrophils in bronchiectasis patients correlated with worse pulmonary function, bacterial colonization, and severe disease [53]. In this context, NE is a key determinant of tissue damage and a potential marker of both disease severity and activity, a predictor of long-term clinical outcomes, and a treatment target. In order to better evaluate the role of NE in bronchiectasis, a systematic search of scientific evidence on NE in bronchiectasis was performed.

Methods

Search methodology

Two investigators (AG and FA) independently performed the search on PubMed and assessed the studies according to predefined criteria. Reference lists of the selected manuscripts were also manually assessed. English language restriction was applied. This systematic revision was conducted according to PRISMA statement [54].

Study selection

We included studies published up to May 15, 2017. Key terms included: ‘bronchiectasis AND neutrophilic elastase OR neutrophil elastase’; ‘bronchiectasis AND protease’; ‘bronchiectasis AND neutrophil elastase inhibitors’; ‘bronchiectasis AND desmosine’. After literature search, titles and abstracts were reviewed by two independent investigators (AG and FA) and in case of disagreement a final decision was taken by the principal investigator (SA). Articles were excluded if: (1) written in languages other than English; (2) they were case reports, case series, or qualitative reviews; (3) NE was not measured; (4) missing bronchiectasis patients; (5) inclusion of CF patients; (6) inclusion of pediatric patients; (7) full-text was unavailable. Full-text was finally obtained for selected papers.

Data extraction and analysis

After the full-text analysis, data of interest from each included paper were extracted. Data of interest included: name of the first author, year of publication, study design, sample size, assay manufacturer, biological matrix, setting, clinical state of patients, quantitative findings and endpoints. Corresponding authors were contacted if data were not present or unclear in the full-text. Given the high degree of heterogeneity across papers taken into account, a meta-analysis was not performed.

Results

The process and results of the search are shown in Fig. 1. The majority of the selected studies were rejected because either they were qualitative reviews or case series (n = 35) or not dealing with bronchiectasis patients (n = 34), or evaluating CF patients (n = 29) /children (n = 5), or their full-text was not available in English (n = 16). A final pool of 31 studies was included in the systematic review, with a total of 2679 patients (Table 2). Selected papers were published from 1984 to 2016, with an elevated frequency in the period 1998–2002 (10/31, 32.3%). The majority had a cross-sectional design (19/31, 61.3%). Only six (19.4%) studies included patients during an acute exacerbation. Measurement of NE was highly heterogeneous, including both commercial kits and in-home assays. Sputum was the most represented biological sample for NE detection. Blood samples were occasionally collected for NE or desmosine measurement [17, 21, 55]. Only one (3.2%) study detected urine desmosine. Most of the selected studies were aimed to assess NE as an inflammatory marker (19/31, 61.3%), while 7 (22.6%) studies investigated NE as a marker of response to treatment and 1 (3.2%) study evaluated NE as a target of therapy.
Table 2
Results of the systematic review
Author and year
Study design
Setting
Clinical stability
Sample size
Assay manufacturer (methods)
Quantitative findings
Biological matrix
Major results
Stockley, 1984 [18]
Cross-sectional
Monocentric, outpatients
Stable state
Exacerbation
34 Bx patients
Elastin-agarose diffusion plate (in-home assay)
NE activity (μg x mL): grade1 19.5; grade2 48.2; grade3 62.9
Sputum
Correlation with sputum macroscopic appearance
Smallman, 1984 [6]
Cross-sectional/Prospective
Monocentric, outpatients
Stable state
8 Bx patients
Elastin-agarose diffusion plate (in-home assay)
NE activity (μg x mL): not quantified but evaluated as present/not present (lower limit of detection 0.8)
Sputum
Inverse correlation with ciliary beat frequency
NE reduction after short-term antibiotic therapy
Fujita, 1992 [60]
Cross-sectional
Monocentric, outpatients
Stable state
9 Bx patients in a cohort of 64 chronic respiratory diseases +15 healthy subjects
Elisa for NE- A1AT complex (in-home assay)
NE-A1AT complex levels (μg x L): 558 ± 198; healthy subjects 122 ± 4
Blood
NE- A1AT complex higher in Bx patients than in healthy subjects
Lloberes, 1992 [63]
Cross- sectional
Monocentric, outpatients
Stable state
26 Bx patients
Elastin-agarose diffusion plate (in-home assay)
NE activity (μg × 100 μL): purulent 7.75 (0–21.5); mucopurulent 1.3 (0–19.5); mucoid 0
Sputum
Correlation with sputum macroscopic appearance
Ip, 1993 [71]
Prospective
Monocentric, outpatients
Exacerbation
12 Bx patients
Spectrophotometry, SLAPN substrate
NE activity (mU × 100 μL): baseline 50.5 ± 17.1; exacerbation before atb 51.8 ± 25.4; 1-week atb −24.3 ± 20.7; 2-week atb −35.1 ± 17.8; 2-week after atb −12.1 ± 35.6; 6-week after atb −32.8 ± 26.8
Sputum
NE reduction after short-term antibiotic therapy
Llewellyn-Jones, 1995 [15]
Prospective
Monocentric, outpatients
Stable state
9 Bx patients +8 healthy subjects
Fluorescence (in-home assay)
NE activity (μg x mL−1): day -7th 4.0 ± 0.46; baseline 4.15 ± 0.69; day 14th 4.25 ± 0.63; day 28th 4.39 ± 0.65; day 63th 4.03 ± 0.67
Blood
No NE reduction after indomethacin
Sepper, 1995 [74]
Cross-sectional
Monocentric, outpatients
Stable state
24 Bx patients +15 healthy subjects
Spectrophotometry, SAAVNA substrate
NE activity (mU x g): mild group 0.21 ± 0.09; moderate group 1.87 ± 1.12; severe group 2.64 ± 1.63; healthy subjects 0.09 ± 0.03
BAL
Correlation with symptoms, exacerbation rate and radiological severity
Nakamura, 1997 [56]
Cross-sectional
Monocentric, outpatients
Stable state
3 Bx patients in a cohort of 8 chronic respiratory diseases +15 healthy subjects
Elisa (in-home assay)
Quantitative findings only reported as figures
BAL
NE higher in Bx than in healthy subjects
Gaga, 1998 [57]
Cross-sectional
Monocentric, outpatients
Stable state
12 Bx patients +11 healthy subjects
Immunocitochemistry (in-home assay)
Neutrophils (cells x mm3): Bx patients 114; healthy subjects 41
Bronchial biopsies
NE higher in Bx than in healthy subjects
Nakamura, 1999 [78]
Prospective
Monocentric, outpatients
Stable state
3 Bx patients in a cohort of 10 chronic respiratory diseases
Elisa (in-home assay)
NE levels (μg x mL−1): baseline 125.5 ± 47.5; 16.8 ± 7.1 after atb
BAL
Ne reduction after 3-month roxithromycin
Ichikawa, 1999 [64]
Cross-sectional
Monocentric, outpatients
Stable state
18 Bx patients in a cohort of 59 chronic respiratory diseases +28 healthy subjects
Spectrofluorometry AMC substrate
NE activity (U): purulent 1239.2 ± 1017.8; mucoid 45.7 ± 115.1
Sputum
Inverse correlation with IgBF levels and sputum purulence
Hill, 2000 [16]
Cross- sectional
Monocentric, outpatients
Stable state
43 Bx patients in a cohort of 160 chronic respiratory diseases
Spectrophotometry
NE levels (nM): mixed flora 0 (0–20); high bacterial load 21 (4–40)
Sputum
Correlation with sputum bacterial load
Schaaf, 2000 [68]
Cross-sectional
Monocentric, outpatients
Stable state
11 Bx patients in a cohort of 66 chronic respiratory diseases +12 healthy subjects
Immunoluminometry
Colorimetry
NE-A1AT complex levels (ng x mL): Bx patients 44 (15–152); healthy subjects 3 (0–15)
BAL
NE higher in Bx than in healthy subjects
Correlation with P. aeruginosa chronic respiratory infection
Tsang, 2000 [75]
Cross-sectional
Monocentric, outpatients
Stable state
30 Bx patients
Spectrophotometry, SAAVNA substrate
NE levels (U x mL): 6.60 ± 3.13
Sputum
Correlation with 24 h sputum volume, sputum inflammatory markers (leukocytes, IL-B and TNFa), radiological severity and spirometry (FEV1, FVC)
Zheng, 2000 [69]
Cross-sectional
Monocentric, outpatients
Stable state
35 Bx patients +18 healthy subjects
Spectrophotometry, SAAVNA substrate
NE levels (Unit x mL−1): P. aeruginosa 222.6 (18.3–267.5); 8.7 (0.6–186.7)
Sputum
Correlation with P. aeruginosa chronic respiratory infection
Viglio, 2000 [21]
Cross-sectional
Monocentric, outpatients
Stable state
13 Bx patients in a cohort of 54 chronic respiratory diseases +24 healthy subjects
HPCE
DES (μg x g creatinine−1): Bx patients 23.39 ± 2.05; CF 23.39 ± 2.02; A1AT deficiency 22.3 ± 7.74; exacerbated COPD 17.15 ± 3.42; stable COPD 14.17 ± 2.33; smokers 11.97 ± 2.75; healthy subjects 9.31 ± 2.75
Urine
Desmosine higher in stable Bx than in other chronic respiratory disease and healthy subjects
Angrill, 2001 [62]
Cross-sectional
Monocentric, outpatients
Stable state
49 Bx patients +9 healthy subjects
Immunoassay (?)
NE levels (μg x L): colonized patients 231 (15–2930); not colonized patients 45 (8–2280); healthy subjects 34 (9–44)
BAL
NE higher in Bx than in healthy subjects
Correlation with neutrophils, IL8 and TNFa in BALF
Correlation with bacterial chronic respiratory infection.
Stockley, 2001 [65]
Cross-sectional
Monocentric, outpatients
Stable state
14 Bx patients +9 smokers
Spectrophotometry, MSAPN substrate
Elisa (in-home assay)
NE levels (μM): sputum colour value-3 0.006 ± 1.0; sputum colour value-7 4.14 ± 1.0
Sputum
Correlation with sputum macroscopic appearance
Correlation with MPO and 24 h sputum volume
Inverse correlation with A1AT
Zheng, 2001 [59]
Cross-sectional
Monocentric, outpatients
Stable state
14 Bx patients +15 healthy subjects
Anti-NE antibodies
Median neutrophils x mm−2: Bx patients 608 (101–1013); healthy subj. 127 (24–630)
Endobronchial biopsies
NE higher in bronchiectatic lamina propria than in healthy subjects
Vandivier, 2002 [58]
Prospective
Monocentric, outpatients
Exacerbation
6 Bx patients in a cohort of 18 chronic respiratory diseases
Spectrophotometry
NE (U x mL): Bx patients 10; CF 3
Sputum
NE higher in Bx
NE as cause of delay in apoptotic cell clearance
Chan, 2003 [12]
Cross-sectional
Monocentric, outpatients
Stable state
10 Bx patients
Spectrophotometry
NE activity range (mM): 0.9–1.2
Sputum
NE not only as free but complexed with polyanionic partners (syndecan-1) and A1AT
Watt, 2004 [72]
Prospective
Monocentric, outpatients
Exacerbation
15 Bx patients +10 healthy subjects
Kinetic assays (?)
NE levels (ng x mL): after atb −73,451 (135,495–12,303)
Sputum
NE reduction after short-term antibiotic therapy
Chan, 2009 [61]
Cross-sectional
Monocentric, outpatients
Stable state
12 Bx patients
Spectrophotometry, MSAPN substrate
ELISA (in-home assay)
NE activity (mM): 1.3 (1–2)
Sputum
NE complexed with syndecan-1
Description of protease/anti-protease balance
Murray, 2010 [77]
RCT
Monocentric, outpatients
Stable state
65 Bx patients
Elastin-agarose diffusion plate (in-home assay)
NE activity gentamicin group vs placebo: (U x mg): baseline 3.6 (0–17.6) vs 4.1 (0–19); 3-month 0 (0–0) vs 0 (0–20.4); 6-month 0 (0–2.9) vs 0 (0–29.1); 9-month 0 (0–7.6) vs 0.9 (0–19.4); 12- month 0 (0–1.8) vs 1.8 (0.17–16); 15-month 7.1 (0–56) vs 2.8 (0.9–18.2)
Sputum
NE reduction after 3-month inhaled gentamicin
Chalmers, 2012 [67]
Prospective
Monocentric, outpatients
Stable state
Exacerbation
385 Bx patients
Elastin-agarose diffusion plate (in-home assay)
NE activity (μg x mL): P. aeruginosa infected 4.9 (1.2–68); P. aeruginosa not infected 1.2 (0.4–147.2); other quantitative findings only reported as figures
Sputum
Correlation with bacterial load and radiological severity
Mandall, 2013 [73]
Prospective
Monocentric, outpatients
Stable state
163 Bx patients
Spectrophotometry
NE activity (U x mL): GERD group 5; non-GERD group 3
Sputum
Correlation with GERD
Stockley, 2013 [55]
RCT
Multicentric, outpatients
Stable state
38 Bx patients
Custom-made immunoassay (in-home assay) for both NE and desmosine
NE activity AZD9668 group vs placebo (μM x L): baseline 24.54 vs 7.28; end of treatment 15.92 vs 7.49
Sputum
Urine
NE reduction after 28-day of oral AZD9668
No difference in desmosine between AZD9668 and placebo
Goeminne, 2014 [66]
Cross-sectional
Monocentric, outpatients
Stable state
49 Bx patients +12 healthy subjects
Enzymatic assay
NE activity (μg x mL): Bx patients 15 (3–23); healthy subject 0.8 (0.4–1.2)
Sputum
Correlation with TNFA, CXCL8 and MMP-9
Inverse correlation with FVC
Liu, 2014 [14]
RCT
Monocentric, outpatients
Stable state
43 Bx patients
ELISA (in-home assay)
Quantitative findings only reported as figures
Sputum
NE reduction after 6-month roxythromicin
Aliberti, 2016 [70]
Prospective
Multicentric, outpatients
Stable state
1145 Bx patients
different assays according to different centers
Quantitative findings only reported as figures
Sputum
Correlation with ‘P. aeruginosa‘cluster and ‘Other chronic infection’ cluster
Chalmers, 2016 [17]
Prospective
 
Stable state
Exacerbation
381 Bx patients
Immunoassay (ProAxsis LTD) Kinetic assay (Sigma Aldrich)
LC-MS for desmosine
NE levels (μg x mL): baseline 0.39 (0–23.5); onset of exacerbation 57.0 (3.3–145); after 14-day atb 0 (0–125.8); 1-month later 1.3 (0–29.9)
Sputum
Serum
Correlation with BSI, MRC, FEV 1 , bacterial load, radiological severity
Correlation with higher exacerbation rate, FEV1 decline, all-cause mortality during 3-year follow up
NE increase during exacerbations and NE reduction after short-term antibiotic therapy
Serum desmosine is associated with age and disease severity
Definitions: atb antibiotics, A1AT alpha-1 anti-trypsin, BAL broncho-alveolar lavage, Bx bronchiectasis, DES desmosine, NE neutrophil elastase, SLAPN succinyl-trialanine-nitroanilide, SAAVNA succinyl-Ala-Ala-Val-nitroanilide, MSAPN methoxysuccinyl-Ala-Ala-Pro-Val-paranitroanilide, HPCE high-performance capillary electrophoresis, LC-MS liquid chromatography-mass spectrometry

Discussion

Clinical relevance of NE role in bronchiectasis has been addressed according to a multidimensional approach: its use as inflammatory biomarkers; potential indicator of severity of disease; predictor of clinical outcomes or response to treatment; target of treatment.

NE as an inflammatory marker

Patients with clinically stable bronchiectasis exhibit a permanent neutrophilic activation in the airways and display higher sputum levels of NE and others inflammatory mediators than healthy subjects [5658]. Because of the presence on NE-positive neutrophils in the lamina propria, the inflammation is assumed to be driven by NE [59].
Fujita and Chan demonstrated in different studies that serum levels of NE-A1AT complex in patients with bronchiectasis were significantly higher in comparison to those of healthy subjects suggesting that lung injury is mainly due to NE overexpression than anti-proteinase system deficiency [12, 60, 61].
Angrill proposed that airway inflammation is permanent and may occur even in the absence of bacterial colonization, as demonstrated by the increase of NE and other inflammatory mediators in the BAL of 49 patients in comparison with healthy subjects [62]. Previous studies reported that purulent sputum was associated with NE concentration, and can be considered a marker for proteolytic and inflammatory activity [18, 6365]. These findings were consistent with recent data reported by Goeminne who demonstrated a strong correlation between sputum purulence and proteolytic enzymes, both of which seem to predict the degree of inflammation and disease severity in bronchiectasis [66].
Moreover, NE has been shown to progressively increase with increasing bacterial load in sputum. Chalmers also reported on a strong association between sputum bacterial load and a cluster of inflammatory mediators in a cohort of 434 bronchiectasis patients [16, 67]. Finally, high levels of NE have been documented in special phenotypes of bronchiectasis patients, such as those with chronic infection with both P. aeruginosa and other bacteria [6870].
As a marker of airway inflammation, NE also increases during exacerbations and decreases after a short course of oral antibiotic treatment [6, 67, 71, 72]. These data suggest that NE may act as marker of airway inflammation both in stable patients and during exacerbations.
A correlation between gastroesophageal reflux disease (GERD) and higher levels of NE is also reported [73].

NE as a predictor of clinical outcomes or response to treatment

The identification of a biomarker to assess disease severity and predict progression and outcomes is still an unmet need in bronchiectasis [51]. In the past decades, several experiences showed that NE is associated with clinical outcomes in different bronchiectasis cohorts [66, 74, 75]. Tsang and coworkers showed a correlation between sputum elastase, radiological involvement, and functional markers in a sample of 30 bronchiectasis patients [75]. However, the heterogeneity of measurement assays for NE among different cohorts and the lack of relevant follow-up periods may have affected the findings. Recently, NE underwent a prospective validation in a large cohort of patients followed over 3 years. This sample was divided into three groups according to negative, intermediate, and high elastase activity levels. NE activity was independently associated with lung function decline over 3 years with a loss of 56.4 ml per year in the high activity group (+20.8 ml in comparison with negative-NE group). In addition, NE was also independently associated with the risk of future exacerbations, shorter time to next exacerbation, and all-cause mortality [17].
In a recent evaluation of clinical phenotypes, the phenotype of patients with chronic infection with P. aeruginosa or other bacteria was associated with both higher NE concentrations and worse clinical outcome [70]. This finding is consistent with a meta-analysis by Finch which showed that P. aeruginosa chronic infection is associated with a threefold increased risk of death and an increase in hospital admissions and exacerbations [76].
Future clinical trials are needed to validate cut-off of NE activity and implement NE as a useful biomarker in the clinical management of bronchiectasis patients.
Local or systemic antibiotic therapy and consequent reduction of bacterial load has been hypothesized to decrease sputum NE levels. Previous findings suggest that short oral courses of antibiotic therapy are effective in reducing NE activity in sputum [6, 67, 71]. Chalmers and Murray proved that long-term nebulized gentamycin significantly reduces free elastase activity in sputum, as well as sputum MPO levels and bacterial load [67, 77].
Roxithromycin treatment may reduce sputum NE in bronchiectasis patients as recently demonstrated by Liu in an open-label 6-month study. These findings demonstrate that macrolide therapy may be beneficial in bronchiectasis and may contribute to the understanding of underlying mechanisms [14, 78].
Selected anti-inflammatory drugs have also been tested as indirect modulators of neutrophilic inflammation in bronchiectasis. In a study by Llewellyn-Jones, a 4-week treatment with indomethacin appeared to have no effect on lung inflammation, as assessed by no significant difference in neutrophil and NE activity within the sputum [15].

NE as a target of treatment

Since NE is a major driver of neutrophilic inflammation in bronchiectasis, it has been proposed to tailor treatment aimed to its direct or indirect inhibition, see Fig. 2.
AZD9668 is a NE inhibitor previously investigated in chronic respiratory diseases, including COPD and CF, with inconclusive results [34, 44, 45]. In a small phase II double-blind trial conducted in 10 different centres, 38 patients were randomized to 4 weeks of oral AZD9668 twice daily. AZD9668 increased FEV1 (by about 100 mL) and slowed vital capacity (by about 130 mL) compared with the placebo group. A trend in reduction of most inflammatory biomarkers was also reported (IL-6 and IL-8). There were no differences between AZD9668 and placebo arms regarding sputum quantity, symptoms, and quality of life. The Authors suggested larger studies of longer duration to assess beneficial effects [55, 79].
BAY 85–8501 is a selective NE inhibitor that has shown efficacy in pre-clinical pharmacological and animal models [80]. A phase II randomized parallel group study assessed the safety and tolerability of 28-day oral administration of BAY 85–8501 in 94 bronchiectasis patients. BAY 85–8501 was generally well tolerated and there were no significant changes in lung function, 24 h sputum quantity, NE activity and concentration, and other biomarkers including urine desmosine [81]. Short duration of treatment and inadequate drug concentration at the target site may be the cause of this lack of efficacy. Regarding this last point, it is reasonable that the inhaled approach instead of the oral administration could allow reaching higher lung concentrations with limited systemic exposure and consequent side effects, thus permitting to counteract that elevated amount of active NE found in the patient’s airways.
With this rationale, CHF6333 is the first inhaled NE inhibitor under development for the treatment of NE-driven lung diseases as dry powder inhaler. In in vitro assays, CHF6333 is highly potent in inhibiting human NE (IC50 = 0.2 nM) with good selectivity against other proteases. In addition, CHF6333 significantly reduces lung neutrophil recruitment induced by cigarette smoke exposure in mice and reduces both lung tissue infection and inflammation when administered intratracheally to P. aeruginosa lung infected rats for 7 days [82].
A phase I trial to investigate safety and pharmacokinetics on healthy male subjects is still in phase of recruitment (clinicaltrial.​gov ID NCT03056326).

Conclusions

Sputum NE has proved useful as an inflammatory marker both in stable state bronchiectasis and during exacerbations and local or systemic antibiotic treatment. NE has also been associated with risk of exacerbation, time to next exacerbation and all-cause mortality. Inhibition of NE as a specific target of treatment in bronchiectasis is at a very early stage. Future interventional studies should evaluate safety and efficacy for new molecules and formulations.

Acknowledgements

Not applicable.

Availability of data and material

Not applicable.

Funding

Not applicable.
Not applicable.
Not applicable.

Competing interests

SA reports grant support from AstraZeneca, Bayer Healthcare, Pfizer Ltd. and GlaxoSmithKline. FB reports research grants from Boehringer Ingelheim, Chiesi, Zambon, and Pfizer, congress lecture fees from Boehringer Ingelheim, Guidotti-Malesci, Menarini, GSK, Chiesi, Pfizer and Novartis, and consultancy fees from AstraZeneca, Menarini, Mundipharma, Novartis, GSK, Teva and Pfizer; DM and MAC are employees of Chiesi Farmaceutici.

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 Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011 Jul 25;11(8):519–31.CrossRefPubMed Mantovani A, Cassatella MA, Costantini C, Jaillon S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011 Jul 25;11(8):519–31.CrossRefPubMed
2.
Zurück zum Zitat Silvestre-Roig C, Hidalgo A, Soehnlein O. Neutrophil heterogeneity: implications for homeostasis and pathogenesis. Blood. 2016 May 5;127(18):2173–81.CrossRefPubMed Silvestre-Roig C, Hidalgo A, Soehnlein O. Neutrophil heterogeneity: implications for homeostasis and pathogenesis. Blood. 2016 May 5;127(18):2173–81.CrossRefPubMed
3.
Zurück zum Zitat Weiss SJ. Tissue destruction by neutrophils. N Engl J Med. 1989 Feb9;320(6):365–376. Weiss SJ. Tissue destruction by neutrophils. N Engl J Med. 1989 Feb9;320(6):365–376.
4.
Zurück zum Zitat Weissmann G, Smolen JE, Korchak HM. Release of inflammatory mediators from stimulated neutrophils. N Engl J Med. 1980 Jul 3;303(1):27–34.CrossRefPubMed Weissmann G, Smolen JE, Korchak HM. Release of inflammatory mediators from stimulated neutrophils. N Engl J Med. 1980 Jul 3;303(1):27–34.CrossRefPubMed
5.
Zurück zum Zitat Tosi MF, Zakem H, Berger M. Neutrophil elastase cleaves C3bi on opsonised pseudomonas as well as CR1 on neutrophils to create a functionally important opsonin receptor mismatch. J Clin Invest. 1990 Jul;86(1):300–8.CrossRefPubMedPubMedCentral Tosi MF, Zakem H, Berger M. Neutrophil elastase cleaves C3bi on opsonised pseudomonas as well as CR1 on neutrophils to create a functionally important opsonin receptor mismatch. J Clin Invest. 1990 Jul;86(1):300–8.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Smallman LA, Hill SL, Stockley RA. Reduction of ciliary beat frequency in vitro by sputum from patients with bronchiectasis: a serine proteinase effect. Thorax. 1984 Sep;39(9):663–7.CrossRefPubMedPubMedCentral Smallman LA, Hill SL, Stockley RA. Reduction of ciliary beat frequency in vitro by sputum from patients with bronchiectasis: a serine proteinase effect. Thorax. 1984 Sep;39(9):663–7.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Amitani R, Wilson R, Rutman A, Read R, Ward C, Burnett D, Stockley RA, Cole PJ. Effects of human neutrophil elastase and Pseudomonas Aeruginosa proteinases on human respiratory epithelium. Am J Respir Cell Mol Biol. 1991 Jan;4(1):26–32.CrossRefPubMed Amitani R, Wilson R, Rutman A, Read R, Ward C, Burnett D, Stockley RA, Cole PJ. Effects of human neutrophil elastase and Pseudomonas Aeruginosa proteinases on human respiratory epithelium. Am J Respir Cell Mol Biol. 1991 Jan;4(1):26–32.CrossRefPubMed
8.
Zurück zum Zitat Cech P, Lehrer RI. Phagolysosomal pH of human neutrophils. Blood. 1984 Jan;63(1):88–95.PubMed Cech P, Lehrer RI. Phagolysosomal pH of human neutrophils. Blood. 1984 Jan;63(1):88–95.PubMed
9.
Zurück zum Zitat Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ. Cell surface-bound elastase and cathepsin G on human neutrophils: a novel, non-oxidative mechanism by which neutrophils focus and preserve catalytic activity of serine proteinases. J Cell Biol. 1995 Nov;131(3):775–89.CrossRefPubMed Owen CA, Campbell MA, Sannes PL, Boukedes SS, Campbell EJ. Cell surface-bound elastase and cathepsin G on human neutrophils: a novel, non-oxidative mechanism by which neutrophils focus and preserve catalytic activity of serine proteinases. J Cell Biol. 1995 Nov;131(3):775–89.CrossRefPubMed
10.
Zurück zum Zitat Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004 Mar 5;303(5663):1532–5.CrossRefPubMed Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, Weinrauch Y, Zychlinsky A. Neutrophil extracellular traps kill bacteria. Science. 2004 Mar 5;303(5663):1532–5.CrossRefPubMed
11.
Zurück zum Zitat Sandhaus RA, Turino G. Neutrophil elastase-mediated lung disease. COPD. 2013 Mar;10(Suppl 1):60–3.CrossRefPubMed Sandhaus RA, Turino G. Neutrophil elastase-mediated lung disease. COPD. 2013 Mar;10(Suppl 1):60–3.CrossRefPubMed
12.
Zurück zum Zitat Chan SC, Shum DK, Ip MS. Sputum sol neutrophil elastase activity in bronchiectasis: differential modulation by syndecan-1. Am J Respir Crit Care Med 2003 Jul 15;168(2):192-8. Epub 2003 Apr 17. Chan SC, Shum DK, Ip MS. Sputum sol neutrophil elastase activity in bronchiectasis: differential modulation by syndecan-1. Am J Respir Crit Care Med 2003 Jul 15;168(2):192-8. Epub 2003 Apr 17.
13.
Zurück zum Zitat Korkmaz B, Moreau T, Gauthier F. Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physiopathological functions. Biochimie 2008 Feb;90(2):227-42. Epub 2007 Oct 25. Korkmaz B, Moreau T, Gauthier F. Neutrophil elastase, proteinase 3 and cathepsin G: physicochemical properties, activity and physiopathological functions. Biochimie 2008 Feb;90(2):227-42. Epub 2007 Oct 25.
14.
Zurück zum Zitat Liu J, Zhong X, He Z, Wei L, Zheng X, Zhang J, Bai J, Zhong W, Zhong D. Effect of low-dose, long-term roxithromycin on airway inflammation and remodeling of stable noncystic fibrosis bronchiectasis. Mediat Inflamm. 2014;2014:708608. Liu J, Zhong X, He Z, Wei L, Zheng X, Zhang J, Bai J, Zhong W, Zhong D. Effect of low-dose, long-term roxithromycin on airway inflammation and remodeling of stable noncystic fibrosis bronchiectasis. Mediat Inflamm. 2014;2014:708608.
15.
Zurück zum Zitat Llewellyn-Jones CG, Johnson MM, Mitchell JL, Pye A, Okafor VC, Hill SL, Stockley RA. In vivo study of indomethacin in bronchiectasis: effect on neutrophil function and lung secretion. Eur Respir J. 1995 Sep;8(9):1479–87.PubMed Llewellyn-Jones CG, Johnson MM, Mitchell JL, Pye A, Okafor VC, Hill SL, Stockley RA. In vivo study of indomethacin in bronchiectasis: effect on neutrophil function and lung secretion. Eur Respir J. 1995 Sep;8(9):1479–87.PubMed
16.
Zurück zum Zitat Hill AT, Campbell EJ, Hill SL, Bayley DL, Stockley RA. Association between airway bacterial load and markers of airway inflammation in patients with stable chronic bronchitis. Am J Med. 2000 Sep;109(4):288–95.CrossRefPubMed Hill AT, Campbell EJ, Hill SL, Bayley DL, Stockley RA. Association between airway bacterial load and markers of airway inflammation in patients with stable chronic bronchitis. Am J Med. 2000 Sep;109(4):288–95.CrossRefPubMed
17.
Zurück zum Zitat Chalmers JD, Moffitt KL, Suarez-Cuartin G, Sibila O, Finch S, Furrie E, Dicker A, Wrobel K, Elborn JS, Walker B, Martin SL, Marshall SE, Huang JT, Fardon TC. Neutrophil Elastase activity is associated with exacerbations and lung function decline in Bronchiectasis. Am J Respir Crit Care Med. 2017 May 15;195(10):1384–93.CrossRefPubMedPubMedCentral Chalmers JD, Moffitt KL, Suarez-Cuartin G, Sibila O, Finch S, Furrie E, Dicker A, Wrobel K, Elborn JS, Walker B, Martin SL, Marshall SE, Huang JT, Fardon TC. Neutrophil Elastase activity is associated with exacerbations and lung function decline in Bronchiectasis. Am J Respir Crit Care Med. 2017 May 15;195(10):1384–93.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Stockley RA, Hill SL, Morrison HM, Starkie CM. Elastolytic activity of sputum and its relation to purulence and to lung function in patients with bronchiectasis. Thorax. 1984 Jun;39(6):408–13.CrossRefPubMedPubMedCentral Stockley RA, Hill SL, Morrison HM, Starkie CM. Elastolytic activity of sputum and its relation to purulence and to lung function in patients with bronchiectasis. Thorax. 1984 Jun;39(6):408–13.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Starcher B, Scott M. Fractionation of urine to allow desmosine analysis by radioimmunoassay. Ann Clin Biochem. 1992 Jan;29(Pt 1):72–8.CrossRefPubMed Starcher B, Scott M. Fractionation of urine to allow desmosine analysis by radioimmunoassay. Ann Clin Biochem. 1992 Jan;29(Pt 1):72–8.CrossRefPubMed
20.
Zurück zum Zitat Albarbarawi O, Barton A, Miller D, McSharry C, Chaudhuri R, Thomson NC, Palmer CN, Devereux G, Huang JT. Characterization and validation of an isotope-dilution LC-MS/MS method for quantification of total desmosine and isodesmosine in plasma and serum. Bioanalysis. 2013 Aug;5(16):1991–2001.CrossRefPubMed Albarbarawi O, Barton A, Miller D, McSharry C, Chaudhuri R, Thomson NC, Palmer CN, Devereux G, Huang JT. Characterization and validation of an isotope-dilution LC-MS/MS method for quantification of total desmosine and isodesmosine in plasma and serum. Bioanalysis. 2013 Aug;5(16):1991–2001.CrossRefPubMed
21.
Zurück zum Zitat Viglio S, Valentini G, Chiarelli L, Zanaboni G, Cetta G, Iadarola P. Micellar electrokinetic chromatography as a complementary method to sodium dodecyl sulfate-polyacrylamide gel electrophoresis for studying limited proteolysis of proteins. Electrophoresis. 1999 Sep;20(12):2400–6.CrossRefPubMed Viglio S, Valentini G, Chiarelli L, Zanaboni G, Cetta G, Iadarola P. Micellar electrokinetic chromatography as a complementary method to sodium dodecyl sulfate-polyacrylamide gel electrophoresis for studying limited proteolysis of proteins. Electrophoresis. 1999 Sep;20(12):2400–6.CrossRefPubMed
22.
Zurück zum Zitat Khan TZ, Wagener JS, Bost T, Martinez J, Accurso FJ, Riches DW. Early pulmonary inflammation in infants with cystic fibrosis. Am J Respir Crit Care Med. 1995 Apr;151(4):1075–82.PubMed Khan TZ, Wagener JS, Bost T, Martinez J, Accurso FJ, Riches DW. Early pulmonary inflammation in infants with cystic fibrosis. Am J Respir Crit Care Med. 1995 Apr;151(4):1075–82.PubMed
23.
Zurück zum Zitat Sly PD, Brennan S, Gangell C, de Klerk N, Murray C, Mott L, Stick SM, Robinson PJ, Robertson CF, Ranganathan SC. Australian respiratory early surveillance team for cystic fibrosis (AREST-CF). Lung disease at diagnosis in infants with cystic fibrosis detected by newborn screening. Am J Respir Crit Care Med. 2009 Jul 15;180(2):146–52.CrossRefPubMed Sly PD, Brennan S, Gangell C, de Klerk N, Murray C, Mott L, Stick SM, Robinson PJ, Robertson CF, Ranganathan SC. Australian respiratory early surveillance team for cystic fibrosis (AREST-CF). Lung disease at diagnosis in infants with cystic fibrosis detected by newborn screening. Am J Respir Crit Care Med. 2009 Jul 15;180(2):146–52.CrossRefPubMed
24.
Zurück zum Zitat Sagel SD, Wagner BD, Anthony MM, Emmett P, Zemanick ET. Sputum biomarkers of inflammation and lung function decline in children with cystic fibrosis. Am J Respir Crit Care Med. 2012;186(9):857–65.CrossRefPubMedPubMedCentral Sagel SD, Wagner BD, Anthony MM, Emmett P, Zemanick ET. Sputum biomarkers of inflammation and lung function decline in children with cystic fibrosis. Am J Respir Crit Care Med. 2012;186(9):857–65.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Park JA, He F, Martin LD, Li Y, Chorley BN, Adler KB. Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C{delta}-mediated mechanism. Am J Pathol. 2005 Sep;167(3):651–61.CrossRefPubMedPubMedCentral Park JA, He F, Martin LD, Li Y, Chorley BN, Adler KB. Human neutrophil elastase induces hypersecretion of mucin from well-differentiated human bronchial epithelial cells in vitro via a protein kinase C{delta}-mediated mechanism. Am J Pathol. 2005 Sep;167(3):651–61.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Rubio F, Cooley J, Accurso FJ, Remold-O'Donnell E. Linkage of neutrophil serine proteases and decreased surfactant protein-a (SP-A) levels in inflammatory lung disease. Thorax. 2004 Apr;59(4):318–23.CrossRefPubMedPubMedCentral Rubio F, Cooley J, Accurso FJ, Remold-O'Donnell E. Linkage of neutrophil serine proteases and decreased surfactant protein-a (SP-A) levels in inflammatory lung disease. Thorax. 2004 Apr;59(4):318–23.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing,whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood. 2000 Oct 15;96(8):2673–81.PubMed Van den Steen PE, Proost P, Wuyts A, Van Damme J, Opdenakker G. Neutrophil gelatinase B potentiates interleukin-8 tenfold by aminoterminal processing,whereas it degrades CTAP-III, PF-4, and GRO-alpha and leaves RANTES and MCP-2 intact. Blood. 2000 Oct 15;96(8):2673–81.PubMed
28.
Zurück zum Zitat Rogan MP, Taggart CC, Greene CM, Murphy PG, O'Neill SJ, McElvaney NG. Loss of microbicidal activity and increased formation of biofilm due to decreased lactoferrin activity in patients with cystic fibrosis. J Infect Dis. 2004 Oct 1;190(7):1245–53.CrossRefPubMed Rogan MP, Taggart CC, Greene CM, Murphy PG, O'Neill SJ, McElvaney NG. Loss of microbicidal activity and increased formation of biofilm due to decreased lactoferrin activity in patients with cystic fibrosis. J Infect Dis. 2004 Oct 1;190(7):1245–53.CrossRefPubMed
29.
Zurück zum Zitat Guyot N, Butler MW, McNally P, Weldon S, Greene CM, Levine RL, O’Neill SJ, Taggart CC, McElvaney NG. Elafin, an elastase-specific inhibitor, is cleaved by its cognate enzyme neutrophil elastase in sputum from individuals with cystic fibrosis. J Biol Chem. 2008 Nov 21;283(47):32377–85.CrossRefPubMedPubMedCentral Guyot N, Butler MW, McNally P, Weldon S, Greene CM, Levine RL, O’Neill SJ, Taggart CC, McElvaney NG. Elafin, an elastase-specific inhibitor, is cleaved by its cognate enzyme neutrophil elastase in sputum from individuals with cystic fibrosis. J Biol Chem. 2008 Nov 21;283(47):32377–85.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Döring G, Frank F, Boudier C, Herbert S, Fleischer B, Bellon G. Cleavage of lymphocyte surface antigens CD2, CD4, and CD8 by polymorphonuclear leukocyte elastase and cathepsin G in patients with cystic fibrosis. J Immunol. 1995 May 1;154(9):4842–50.PubMed Döring G, Frank F, Boudier C, Herbert S, Fleischer B, Bellon G. Cleavage of lymphocyte surface antigens CD2, CD4, and CD8 by polymorphonuclear leukocyte elastase and cathepsin G in patients with cystic fibrosis. J Immunol. 1995 May 1;154(9):4842–50.PubMed
31.
Zurück zum Zitat Hartl D, Latzin P, Hordijk P, Marcos V, Rudolph C, Woischnik M, Krauss-Etschmann S, Koller B, Reinhardt D, Roscher AA, Roos D, Griese M. Cleavage of CXCR1 on neutrophils disables bacterial killing in cystic fibrosis in lung disease. Nat Med. 2007 Dec;13(12):1423–30.CrossRefPubMed Hartl D, Latzin P, Hordijk P, Marcos V, Rudolph C, Woischnik M, Krauss-Etschmann S, Koller B, Reinhardt D, Roscher AA, Roos D, Griese M. Cleavage of CXCR1 on neutrophils disables bacterial killing in cystic fibrosis in lung disease. Nat Med. 2007 Dec;13(12):1423–30.CrossRefPubMed
32.
Zurück zum Zitat Gentzsch M, Dang H, Dang Y, Garcia-Caballero A, Suchindran H, Boucher RC, Stutts MJ. The cystic fibrosis transmembrane conductance regulator impende proteolytic stimulation of the epithelial Na+ channel. J Biol Chem. 2010 Oct 15;285(42):32227–32.CrossRefPubMedPubMedCentral Gentzsch M, Dang H, Dang Y, Garcia-Caballero A, Suchindran H, Boucher RC, Stutts MJ. The cystic fibrosis transmembrane conductance regulator impende proteolytic stimulation of the epithelial Na+ channel. J Biol Chem. 2010 Oct 15;285(42):32227–32.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Le Gars M, Descamps D, Roussel D, Saussereau E, Guillot L, Ruffin M, Tabary O, Hong SS, Boulanger P, Paulais M, Malleret L, Belaaouaj A, Edelman A, Huerre M, Chignard M, Sallenave JM. Neutrophil elastase degrades cystic fibrosis transmembrane conductance regulator via calpains and disables channel function in vitro and in vivo. Am J Respir Crit Care Med. 2013 Jan 15;187(2):170–9.CrossRefPubMed Le Gars M, Descamps D, Roussel D, Saussereau E, Guillot L, Ruffin M, Tabary O, Hong SS, Boulanger P, Paulais M, Malleret L, Belaaouaj A, Edelman A, Huerre M, Chignard M, Sallenave JM. Neutrophil elastase degrades cystic fibrosis transmembrane conductance regulator via calpains and disables channel function in vitro and in vivo. Am J Respir Crit Care Med. 2013 Jan 15;187(2):170–9.CrossRefPubMed
34.
Zurück zum Zitat Elborn JS, Perrett J, Forsman-Semb K, Marks-Konczalik J, Gunawardena K, Entwistle N. Efficacy, safety and effect on biomarkers of AZD9668 in cystic fibrosis. Eur Respir J. 2012 Oct;40(4):969–76.CrossRefPubMed Elborn JS, Perrett J, Forsman-Semb K, Marks-Konczalik J, Gunawardena K, Entwistle N. Efficacy, safety and effect on biomarkers of AZD9668 in cystic fibrosis. Eur Respir J. 2012 Oct;40(4):969–76.CrossRefPubMed
35.
Zurück zum Zitat Griese M, Latzin P, Kappler M, Weckerle K, Heinzlmaier T, Bernhardt T, Hartl D. alpha1-antitrypsin inhalation reduces airway inflammation in cystic fibrosis patients. Eur Respir J. 2007 Feb;29(2):240–50.CrossRefPubMed Griese M, Latzin P, Kappler M, Weckerle K, Heinzlmaier T, Bernhardt T, Hartl D. alpha1-antitrypsin inhalation reduces airway inflammation in cystic fibrosis patients. Eur Respir J. 2007 Feb;29(2):240–50.CrossRefPubMed
36.
Zurück zum Zitat Stănescu D, Sanna A, Veriter C, Kostianev S, Calcagni PG, Fabbri LM, Maestrelli P. Airways obstruction, chronic expectoration, and rapid decline of FEV1 in smokers are associated with increased levels of sputum neutrophils. Thorax. 1996 Mar;51(3):267–71.CrossRefPubMedPubMedCentral Stănescu D, Sanna A, Veriter C, Kostianev S, Calcagni PG, Fabbri LM, Maestrelli P. Airways obstruction, chronic expectoration, and rapid decline of FEV1 in smokers are associated with increased levels of sputum neutrophils. Thorax. 1996 Mar;51(3):267–71.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Barnes PJ. The cytokine network in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2009 Dec;41(6):631–8.CrossRefPubMed Barnes PJ. The cytokine network in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2009 Dec;41(6):631–8.CrossRefPubMed
38.
Zurück zum Zitat Richens TR, Linderman DJ, Horstmann SA, Lambert C, Xiao YQ, Keith RL, Boé DM, Morimoto K, Bowler RP, Day BJ, Janssen WJ, Henson PM, Vandivier RW. Cigarette smoke impairs clearance of apoptotic cells through oxidant-dependent activation of RhoA. Am J Respir Crit Care Med. 2009 Jun 1;179(11):1011–21.CrossRefPubMedPubMedCentral Richens TR, Linderman DJ, Horstmann SA, Lambert C, Xiao YQ, Keith RL, Boé DM, Morimoto K, Bowler RP, Day BJ, Janssen WJ, Henson PM, Vandivier RW. Cigarette smoke impairs clearance of apoptotic cells through oxidant-dependent activation of RhoA. Am J Respir Crit Care Med. 2009 Jun 1;179(11):1011–21.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Damiano VV, Tsang A, Kucich U, Abrams WR, Rosenbloom J, Kimbel P, Fallahnejad M, Weinbaum G. Immunolocalization of elastase in human emphysematous lungs. J Clin Invest. 1986 Aug;78(2):482–93.CrossRefPubMedPubMedCentral Damiano VV, Tsang A, Kucich U, Abrams WR, Rosenbloom J, Kimbel P, Fallahnejad M, Weinbaum G. Immunolocalization of elastase in human emphysematous lungs. J Clin Invest. 1986 Aug;78(2):482–93.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Lungarella G, Cavarra E, Lucattelli M, Martorana PA. The dual role of neutrophil elastase in lung destruction and repair. Int J Biochem Cell Biol. 2008;40(6–7):1287–96.CrossRefPubMed Lungarella G, Cavarra E, Lucattelli M, Martorana PA. The dual role of neutrophil elastase in lung destruction and repair. Int J Biochem Cell Biol. 2008;40(6–7):1287–96.CrossRefPubMed
41.
Zurück zum Zitat Shapiro SD, Goldstein NM, Houghton AM, Kobayashi DK, Kelley D, Belaaouaj A. Neutrophil elastase contributes to cigarette smoke-induced emphysema in mice. Am J Pathol. 2003 Dec;163(6):2329–35.CrossRefPubMedPubMedCentral Shapiro SD, Goldstein NM, Houghton AM, Kobayashi DK, Kelley D, Belaaouaj A. Neutrophil elastase contributes to cigarette smoke-induced emphysema in mice. Am J Pathol. 2003 Dec;163(6):2329–35.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Paone G, Conti V, Vestri A, Leone A, Puglisi G, Benassi F, Brunetti G, Schmid G, Cammarella I, Terzano C. Analysis of sputum markers in the evaluation of lung inflammation and functional impairment in symptomatic smokers and COPD patients. Dis Markers. 2011;31(2):91–100.CrossRefPubMedPubMedCentral Paone G, Conti V, Vestri A, Leone A, Puglisi G, Benassi F, Brunetti G, Schmid G, Cammarella I, Terzano C. Analysis of sputum markers in the evaluation of lung inflammation and functional impairment in symptomatic smokers and COPD patients. Dis Markers. 2011;31(2):91–100.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Stevens T, Ekholm K, Gränse M, Lindahl M, Kozma V, Jungar C, Ottosson T, Falk-Håkansson H, Churg A, Wright JL, Lal H, Sanfridson A. AZD9668: pharmacological characterization of a novel oral inhibitor of neutrophil elastase. J Pharmacol Exp Ther. 2011 Oct;339(1):313–20.CrossRefPubMed Stevens T, Ekholm K, Gränse M, Lindahl M, Kozma V, Jungar C, Ottosson T, Falk-Håkansson H, Churg A, Wright JL, Lal H, Sanfridson A. AZD9668: pharmacological characterization of a novel oral inhibitor of neutrophil elastase. J Pharmacol Exp Ther. 2011 Oct;339(1):313–20.CrossRefPubMed
44.
Zurück zum Zitat Kuna P, Jenkins M, O'Brien CD, Fahy WA. AZD9668, a neutrophil elastase inhibitor, plus ongoing budesonide/formoterol in patients with COPD. Respir Med. 2012 Apr;106(4):531–9.CrossRefPubMed Kuna P, Jenkins M, O'Brien CD, Fahy WA. AZD9668, a neutrophil elastase inhibitor, plus ongoing budesonide/formoterol in patients with COPD. Respir Med. 2012 Apr;106(4):531–9.CrossRefPubMed
45.
Zurück zum Zitat Vogelmeier C, Aquino TO, O'Brien CD, Perrett J, Gunawardena KA. A randomised, placebo-controlled, dose-finding study of AZD9668, an oral inhibitor of neutrophil elastase, in patients with chronic obstructive pulmonary disease treated with tiotropium. COPD. 2012 Apr;9(2):111–20.CrossRefPubMed Vogelmeier C, Aquino TO, O'Brien CD, Perrett J, Gunawardena KA. A randomised, placebo-controlled, dose-finding study of AZD9668, an oral inhibitor of neutrophil elastase, in patients with chronic obstructive pulmonary disease treated with tiotropium. COPD. 2012 Apr;9(2):111–20.CrossRefPubMed
46.
Zurück zum Zitat Chapman KR, Burdon JG, Piitulainen E, Sandhaus RA, Seersholm N, Stocks JM, Stoel BC, Huang L, Yao Z, Edelman JM, NG ME. RAPID trial study group. Intravenous augmentation treatment and lung density in severe α1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet. 2015 Jul 25;386(9991):360–8.CrossRefPubMed Chapman KR, Burdon JG, Piitulainen E, Sandhaus RA, Seersholm N, Stocks JM, Stoel BC, Huang L, Yao Z, Edelman JM, NG ME. RAPID trial study group. Intravenous augmentation treatment and lung density in severe α1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet. 2015 Jul 25;386(9991):360–8.CrossRefPubMed
47.
Zurück zum Zitat Gibson PG, Simpson JL, Saltos N. Heterogeneity of airway inflammation in persistent asthma: evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest. 2001 May;119(5):1329–36.CrossRefPubMed Gibson PG, Simpson JL, Saltos N. Heterogeneity of airway inflammation in persistent asthma: evidence of neutrophilic inflammation and increased sputum interleukin-8. Chest. 2001 May;119(5):1329–36.CrossRefPubMed
48.
Zurück zum Zitat Louis R, Djukanovic R. Is the neutrophil a worthy target in severe asthma and chronic obstructive pulmonary disease? Clin Exp Allergy. 2006 May;36(5):563–7.CrossRefPubMed Louis R, Djukanovic R. Is the neutrophil a worthy target in severe asthma and chronic obstructive pulmonary disease? Clin Exp Allergy. 2006 May;36(5):563–7.CrossRefPubMed
49.
Zurück zum Zitat Simpson JL, Scott RJ, Boyle MJ, Gibson PG. Differential proteolytic enzyme activity in eosinophilic and neutrophilic asthma. Am J Respir Crit Care Med. 2005 Sep 1;172(5):559–65.CrossRefPubMed Simpson JL, Scott RJ, Boyle MJ, Gibson PG. Differential proteolytic enzyme activity in eosinophilic and neutrophilic asthma. Am J Respir Crit Care Med. 2005 Sep 1;172(5):559–65.CrossRefPubMed
50.
Zurück zum Zitat Chalmers JD, Aliberti S, Blasi F. Management of bronchiectasis in adults. Eur Respir J. 2015 May;45(5):1446–62.CrossRefPubMed Chalmers JD, Aliberti S, Blasi F. Management of bronchiectasis in adults. Eur Respir J. 2015 May;45(5):1446–62.CrossRefPubMed
51.
Zurück zum Zitat Aliberti S, Masefield S, Polverino E, De Soyza A, Loebinger MR, Menendez R, Ringshausen FC, Vendrell M, Powell P, Chalmers JD, EMBARC Study Group. Research priorities in bronchiectasis: a consensus statement from the EMBARC clinical research collaboration. Eur Respir J. 2016 Sep;48(3):632–47.CrossRefPubMed Aliberti S, Masefield S, Polverino E, De Soyza A, Loebinger MR, Menendez R, Ringshausen FC, Vendrell M, Powell P, Chalmers JD, EMBARC Study Group. Research priorities in bronchiectasis: a consensus statement from the EMBARC clinical research collaboration. Eur Respir J. 2016 Sep;48(3):632–47.CrossRefPubMed
52.
Zurück zum Zitat Amati F, Franceschi E, Gramegna A, Chalmers JD, Aliberti S. Investigating the etiology of Bronchiectasis: you do not find what you do not look for. Respiration. 2017;93(3):228–9.CrossRefPubMed Amati F, Franceschi E, Gramegna A, Chalmers JD, Aliberti S. Investigating the etiology of Bronchiectasis: you do not find what you do not look for. Respiration. 2017;93(3):228–9.CrossRefPubMed
53.
Zurück zum Zitat Dente FL, Bilotta M, Bartoli ML, et al. Neutrophilic bronchial inflammation correlates with clinical and functional findings in patients with noncystic fibrosis Bronchiectasis. Mediat Inflamm. 2015;2015:642503.CrossRef Dente FL, Bilotta M, Bartoli ML, et al. Neutrophilic bronchial inflammation correlates with clinical and functional findings in patients with noncystic fibrosis Bronchiectasis. Mediat Inflamm. 2015;2015:642503.CrossRef
54.
Zurück zum Zitat Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med 2009 Aug 18;151(4):264-9, W64. Moher D, Liberati A, Tetzlaff J, Altman DG; PRISMA group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med 2009 Aug 18;151(4):264-9, W64.
55.
Zurück zum Zitat Stockley R, De Soyza A, Gunawardena K, Perrett J, Forsman-Semb K, Entwistle N, Snell N. Phase II study of a neutrophil elastase inhibitor (AZD9668) in patients with bronchiectasis. Respir Med. 2013 Apr;107(4):524–33.CrossRefPubMed Stockley R, De Soyza A, Gunawardena K, Perrett J, Forsman-Semb K, Entwistle N, Snell N. Phase II study of a neutrophil elastase inhibitor (AZD9668) in patients with bronchiectasis. Respir Med. 2013 Apr;107(4):524–33.CrossRefPubMed
56.
Zurück zum Zitat Nakamura H, Abe S, Shibata Y, Yuki H, Suzuki H, Saito H, Sata M, Kato S, Tomoike H. Elevated levels of cytokeratin 19 in the bronchoalveolar lavage fluid of patients with chronic airway inflammatory diseases--a specific marker for bronchial epithelial injury. Am J Respir Crit Care Med. 1997 Apr;155(4):1217–21.CrossRefPubMed Nakamura H, Abe S, Shibata Y, Yuki H, Suzuki H, Saito H, Sata M, Kato S, Tomoike H. Elevated levels of cytokeratin 19 in the bronchoalveolar lavage fluid of patients with chronic airway inflammatory diseases--a specific marker for bronchial epithelial injury. Am J Respir Crit Care Med. 1997 Apr;155(4):1217–21.CrossRefPubMed
57.
Zurück zum Zitat Gaga M, Bentley AM, Humbert M, Barkans J, O'Brien F, Wathen CG, Kay AB, Durham SR. Increases in CD4+ T lymphocytes, macrophages, neutrophils and interleukin 8 positive cells in the airways of patients with bronchiectasis. Thorax. 1998 Aug;53(8):685–91.CrossRefPubMedPubMedCentral Gaga M, Bentley AM, Humbert M, Barkans J, O'Brien F, Wathen CG, Kay AB, Durham SR. Increases in CD4+ T lymphocytes, macrophages, neutrophils and interleukin 8 positive cells in the airways of patients with bronchiectasis. Thorax. 1998 Aug;53(8):685–91.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Vandivier RW, Fadok VA, Hoffmann PR, Bratton DL, Penvari C, Brown KK, Brain JD, Accurso FJ, Henson PM. Elastase-mediated phosphatidylserine receptor cleavage impairs apoptotic cell clearance in cystic fibrosis and bronchiectasis. J Clin Invest. 2002 Mar;109(5):661–70.CrossRefPubMedPubMedCentral Vandivier RW, Fadok VA, Hoffmann PR, Bratton DL, Penvari C, Brown KK, Brain JD, Accurso FJ, Henson PM. Elastase-mediated phosphatidylserine receptor cleavage impairs apoptotic cell clearance in cystic fibrosis and bronchiectasis. J Clin Invest. 2002 Mar;109(5):661–70.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Zheng L, Shum H, Tipoe GL, Leung R, Lam WK, Ooi GC, Tsang KW. Macrophages, neutrophils and tumour necrosis factor-alpha expression in bronchiectatic airways in vivo. Respir Med. 2001 Oct;95(10):792–8.CrossRefPubMed Zheng L, Shum H, Tipoe GL, Leung R, Lam WK, Ooi GC, Tsang KW. Macrophages, neutrophils and tumour necrosis factor-alpha expression in bronchiectatic airways in vivo. Respir Med. 2001 Oct;95(10):792–8.CrossRefPubMed
60.
Zurück zum Zitat Fujita J, Nakamura H, Yamagishi Y, Yamaji Y, Shiotani T, Irino S. Elevation of plasma truncated elastase alpha 1-proteinase inhibitor complexes in patients with inflammatory lung diseases. Chest. 1992 Jul;102(1):129–34.CrossRefPubMed Fujita J, Nakamura H, Yamagishi Y, Yamaji Y, Shiotani T, Irino S. Elevation of plasma truncated elastase alpha 1-proteinase inhibitor complexes in patients with inflammatory lung diseases. Chest. 1992 Jul;102(1):129–34.CrossRefPubMed
61.
Zurück zum Zitat Chan SC, Leung VO, Ip MS, Shum DK. Shed syndecan-1 restricts neutrophil elastase from alpha1-antitrypsin in neutrophilic airway inflammation. Am J Respir Cell Mol Biol. 2009 Nov;41(5):620–8.CrossRefPubMed Chan SC, Leung VO, Ip MS, Shum DK. Shed syndecan-1 restricts neutrophil elastase from alpha1-antitrypsin in neutrophilic airway inflammation. Am J Respir Cell Mol Biol. 2009 Nov;41(5):620–8.CrossRefPubMed
62.
Zurück zum Zitat Angrill J, Agustí C, De Celis R, Filella X, Rañó A, Elena M, De La Bellacasa JP, Xaubet A, Torres A. Bronchial inflammation and colonization in patients with clinically stable bronchiectasis. Am J Respir Crit Care Med. 2001 Nov 1;164(9):1628–32.CrossRefPubMed Angrill J, Agustí C, De Celis R, Filella X, Rañó A, Elena M, De La Bellacasa JP, Xaubet A, Torres A. Bronchial inflammation and colonization in patients with clinically stable bronchiectasis. Am J Respir Crit Care Med. 2001 Nov 1;164(9):1628–32.CrossRefPubMed
63.
Zurück zum Zitat Lloberes P, Montserrat E, Montserrat JM, Picado C. Sputum sol phase proteins and elastase activity in patients with clinically stable bronchiectasis. Thorax. 1992 Feb;47(2):88–92.CrossRefPubMedPubMedCentral Lloberes P, Montserrat E, Montserrat JM, Picado C. Sputum sol phase proteins and elastase activity in patients with clinically stable bronchiectasis. Thorax. 1992 Feb;47(2):88–92.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Ichikawa W, Ogushi F, Tani K, Maniwa K, Kamada M, Ohmoto Y, Sakatani M, Sone S. Characterization of immunoglobulin binding factor in sputum from patients with chronic airway diseases. Respirology. 1999 Dec;4(4):375–81.CrossRefPubMed Ichikawa W, Ogushi F, Tani K, Maniwa K, Kamada M, Ohmoto Y, Sakatani M, Sone S. Characterization of immunoglobulin binding factor in sputum from patients with chronic airway diseases. Respirology. 1999 Dec;4(4):375–81.CrossRefPubMed
65.
Zurück zum Zitat Stockley RA, Bayley D, Hill SL, Hill AT, Crooks S, Campbell EJ. Assessment of airway neutrophils by sputum colour: correlation with airways inflammation. Thorax. 2001 May;56(5):366–72.CrossRefPubMedPubMedCentral Stockley RA, Bayley D, Hill SL, Hill AT, Crooks S, Campbell EJ. Assessment of airway neutrophils by sputum colour: correlation with airways inflammation. Thorax. 2001 May;56(5):366–72.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Goeminne PC, Vandooren J, Moelants EA, Decraene A, Rabaey E, Pauwels A, Seys S, Opdenakker G, Proost P, Dupont LJ. The sputum colour chart as a predictor of lung inflammation, proteolysis and damage in non-cystic fibrosis bronchiectasis: a case-control analysis. Respirology. 2014 Feb;19(2):203–10.CrossRefPubMed Goeminne PC, Vandooren J, Moelants EA, Decraene A, Rabaey E, Pauwels A, Seys S, Opdenakker G, Proost P, Dupont LJ. The sputum colour chart as a predictor of lung inflammation, proteolysis and damage in non-cystic fibrosis bronchiectasis: a case-control analysis. Respirology. 2014 Feb;19(2):203–10.CrossRefPubMed
67.
Zurück zum Zitat Chalmers JD, Smith MP, McHugh BJ, Doherty C, Govan JR, Hill AT. Short- and long term antibiotic treatment reduces airway and systemic inflammation in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2012 Oct 1;186(7):657–65.CrossRefPubMed Chalmers JD, Smith MP, McHugh BJ, Doherty C, Govan JR, Hill AT. Short- and long term antibiotic treatment reduces airway and systemic inflammation in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2012 Oct 1;186(7):657–65.CrossRefPubMed
68.
Zurück zum Zitat Schaaf B, Wieghorst A, Aries SP, Dalhoff K, Braun J. Neutrophil inflammation and activation in bronchiectasis: comparison with pneumonia and idiopathic pulmonary fibrosis. Respiration. 2000;67(1):52–9.CrossRefPubMed Schaaf B, Wieghorst A, Aries SP, Dalhoff K, Braun J. Neutrophil inflammation and activation in bronchiectasis: comparison with pneumonia and idiopathic pulmonary fibrosis. Respiration. 2000;67(1):52–9.CrossRefPubMed
69.
Zurück zum Zitat Zheng L, Tipoe G, Lam WK, Ho JC, Shum I, Ooi GC, Leung R, Tsang KW. Endothelin-1 in stable bronchiectasis. Eur Respir J. 2000 Jul;16(1):146–9.CrossRefPubMed Zheng L, Tipoe G, Lam WK, Ho JC, Shum I, Ooi GC, Leung R, Tsang KW. Endothelin-1 in stable bronchiectasis. Eur Respir J. 2000 Jul;16(1):146–9.CrossRefPubMed
70.
Zurück zum Zitat Aliberti S, Lonni S, Dore S, McDonnell MJ, Goeminne PC, Dimakou K, Fardon TC, Rutherford R, Pesci A, Restrepo MI, Sotgiu G, Chalmers JD. Clinical phenotypes in adult patients with bronchiectasis. Eur Respir J. 2016 Apr;47(4):1113–22.CrossRefPubMed Aliberti S, Lonni S, Dore S, McDonnell MJ, Goeminne PC, Dimakou K, Fardon TC, Rutherford R, Pesci A, Restrepo MI, Sotgiu G, Chalmers JD. Clinical phenotypes in adult patients with bronchiectasis. Eur Respir J. 2016 Apr;47(4):1113–22.CrossRefPubMed
71.
Zurück zum Zitat Ip M, Shum D, Lauder I, Lam WK, So SY. Effect of antibiotics on sputum inflammatory contents in acute exacerbations of bronchiectasis. Respir Med. 1993 Aug;87(6):449–54.CrossRefPubMed Ip M, Shum D, Lauder I, Lam WK, So SY. Effect of antibiotics on sputum inflammatory contents in acute exacerbations of bronchiectasis. Respir Med. 1993 Aug;87(6):449–54.CrossRefPubMed
72.
Zurück zum Zitat Watt AP, Brown V, Courtney J, Kelly M, Garske L, Elborn JS, Ennis M. Neutrophil apoptosis, proinflammatory mediators and cell counts in bronchiectasis. Thorax. 2004 Mar;59(3):231–6.CrossRefPubMedPubMedCentral Watt AP, Brown V, Courtney J, Kelly M, Garske L, Elborn JS, Ennis M. Neutrophil apoptosis, proinflammatory mediators and cell counts in bronchiectasis. Thorax. 2004 Mar;59(3):231–6.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Mandal P, Morice AH, Chalmers JD, Hill AT. Symptoms of airway reflux predict exacerbations and quality of life in bronchiectasis. Respir Med. 2013 Jul;107(7):1008–13.CrossRefPubMed Mandal P, Morice AH, Chalmers JD, Hill AT. Symptoms of airway reflux predict exacerbations and quality of life in bronchiectasis. Respir Med. 2013 Jul;107(7):1008–13.CrossRefPubMed
74.
Zurück zum Zitat Sepper R, Konttinen YT, Ingman T, Sorsa T. Presence, activities, and molecular forms of cathepsin G, elastase, alpha 1-antitrypsin, and alpha 1-antichymotrypsin in bronchiectasis. J Clin Immunol. 1995 Jan;15(1):27–34.CrossRefPubMed Sepper R, Konttinen YT, Ingman T, Sorsa T. Presence, activities, and molecular forms of cathepsin G, elastase, alpha 1-antitrypsin, and alpha 1-antichymotrypsin in bronchiectasis. J Clin Immunol. 1995 Jan;15(1):27–34.CrossRefPubMed
75.
Zurück zum Zitat Tsang KW, Chan K, Ho P, Zheng L, Ooi GC, Ho JC, Lam W. Sputum elastase in steady-state bronchiectasis. Chest. 2000 Feb;117(2):420–6.CrossRefPubMed Tsang KW, Chan K, Ho P, Zheng L, Ooi GC, Ho JC, Lam W. Sputum elastase in steady-state bronchiectasis. Chest. 2000 Feb;117(2):420–6.CrossRefPubMed
76.
Zurück zum Zitat Finch S, McDonnell MJ, Abo-Leyah H, Aliberti S, Chalmers JD. A comprehensive analysis of the impact of Pseudomonas Aeruginosa colonization on prognosis in adult Bronchiectasis. Ann Am Thorac Soc. 2015 Nov;12(11):1602–11.PubMed Finch S, McDonnell MJ, Abo-Leyah H, Aliberti S, Chalmers JD. A comprehensive analysis of the impact of Pseudomonas Aeruginosa colonization on prognosis in adult Bronchiectasis. Ann Am Thorac Soc. 2015 Nov;12(11):1602–11.PubMed
77.
Zurück zum Zitat Murray MP, Govan JR, Doherty CJ, Simpson AJ, Wilkinson TS, Chalmers JD, Greening AP, Haslett C, Hill AT. A randomized controlled trial of nebulized gentamicin in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2011 Feb 15;183(4):491–9.CrossRefPubMed Murray MP, Govan JR, Doherty CJ, Simpson AJ, Wilkinson TS, Chalmers JD, Greening AP, Haslett C, Hill AT. A randomized controlled trial of nebulized gentamicin in non-cystic fibrosis bronchiectasis. Am J Respir Crit Care Med. 2011 Feb 15;183(4):491–9.CrossRefPubMed
78.
Zurück zum Zitat Nakamura H, Fujishima S, Inoue T, Ohkubo Y, Soejima K, Waki Y, Mori M, Urano T, Sakamaki F, Tasaka S, Ishizaka A, Kanazawa M, Yamaguchi K. Clinical and immunoregulatory effects of roxithromycin therapy for chronic respiratory tract infection. Eur Respir J. 1999 Jun;13(6):1371–9.CrossRefPubMed Nakamura H, Fujishima S, Inoue T, Ohkubo Y, Soejima K, Waki Y, Mori M, Urano T, Sakamaki F, Tasaka S, Ishizaka A, Kanazawa M, Yamaguchi K. Clinical and immunoregulatory effects of roxithromycin therapy for chronic respiratory tract infection. Eur Respir J. 1999 Jun;13(6):1371–9.CrossRefPubMed
79.
Zurück zum Zitat Gunawardena KA, Gullstrand H, Perrett J. Pharmacokinetics and safety of AZD9668, an oral neutrophil elastase inhibitor, in healthy volunteers and patients with COPD. Int J Clin Pharmacol Ther. 2013 Apr;51(4):288–304.CrossRefPubMed Gunawardena KA, Gullstrand H, Perrett J. Pharmacokinetics and safety of AZD9668, an oral neutrophil elastase inhibitor, in healthy volunteers and patients with COPD. Int J Clin Pharmacol Ther. 2013 Apr;51(4):288–304.CrossRefPubMed
80.
Zurück zum Zitat von Nussbaum F, Li VM, Allerheiligen S, Anlauf S, Bärfacker L, Bechem M, Delbeck M, Fitzgerald MF, Gerisch M, Gielen-Haertwig H, Haning H, Karthaus D, Lang D, Lustig K, Meibom D, Mittendorf J, Rosentreter U, Schäfer M, Schäfer S, Schamberger J, Telan LA, Tersteegen A. Freezing the bioactive conformation to boost potency: the identification of BAY 85-8501, a selective and potent inhibitor of human Neutrophil Elastase for pulmonary diseases. ChemMedChem. 2015 Jul;10(7):1163–73.CrossRefPubMedPubMedCentral von Nussbaum F, Li VM, Allerheiligen S, Anlauf S, Bärfacker L, Bechem M, Delbeck M, Fitzgerald MF, Gerisch M, Gielen-Haertwig H, Haning H, Karthaus D, Lang D, Lustig K, Meibom D, Mittendorf J, Rosentreter U, Schäfer M, Schäfer S, Schamberger J, Telan LA, Tersteegen A. Freezing the bioactive conformation to boost potency: the identification of BAY 85-8501, a selective and potent inhibitor of human Neutrophil Elastase for pulmonary diseases. ChemMedChem. 2015 Jul;10(7):1163–73.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Watz H, Pedersen F, Kirsten A, Nagelschmitz J, Bandel TJ, Schwers S, Rabe K. Safety and tolerability of the NE inhibitor BAY 85-8501 in patients with non-CF bronchiectasis. ERS International Congress 2016. Watz H, Pedersen F, Kirsten A, Nagelschmitz J, Bandel TJ, Schwers S, Rabe K. Safety and tolerability of the NE inhibitor BAY 85-8501 in patients with non-CF bronchiectasis. ERS International Congress 2016.
82.
Zurück zum Zitat Miglietta D, Carnini C, Puviani V, Finch H, Fox C, Fitzgerald M, Patacchini R, Civelli M, Villetti G. Pharmacological characterization of CHF6333, a novel potent inhaled inhibitor of neutrophil elastase. ERS International Congress 2016. Miglietta D, Carnini C, Puviani V, Finch H, Fox C, Fitzgerald M, Patacchini R, Civelli M, Villetti G. Pharmacological characterization of CHF6333, a novel potent inhaled inhibitor of neutrophil elastase. ERS International Congress 2016.
Metadaten
Titel
Neutrophil elastase in bronchiectasis
verfasst von
Andrea Gramegna
Francesco Amati
Leonardo Terranova
Giovanni Sotgiu
Paolo Tarsia
Daniela Miglietta
Maria Adelaide Calderazzo
Stefano Aliberti
Francesco Blasi
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2017
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-017-0691-x

Weitere Artikel der Ausgabe 1/2017

Respiratory Research 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.