Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2024

Open Access 01.12.2024 | Research

Clinical outcomes of long-term inhaled combination therapies in patients with bronchiectasis and airflow obstruction

verfasst von: Hyo Jin Lee, Jung-Kyu Lee, Tae Yeon Park, Eun Young Heo, Deog Kyeom Kim, Hyun Woo Lee

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2024

Abstract

Background and objectives

Few studies have reported which inhaled combination therapy, either bronchodilators and/or inhaled corticosteroids (ICSs), is beneficial in patients with bronchiectasis and airflow obstruction. Our study compared the efficacy and safety among different inhaled combination therapies in patients with bronchiectasis and airflow obstruction.

Methods

Our retrospective study analyzed the patients with forced expiratory volume in 1 s (FEV1)/forced vital capacity < 0.7 and radiologically confirmed bronchiectasis in chest computed tomography between January 2005 and December 2021. The eligible patients underwent baseline and follow-up spirometric assessments. The primary endpoint was the development of a moderate-to-severe exacerbation. The secondary endpoints were the change in the annual FEV1 and the adverse events. Subgroup analyses were performed according to the blood eosinophil count (BEC).

Results

Among 179 patients, the ICS/long-acting beta-agonist (LABA)/long-acting muscarinic antagonist (LAMA), ICS/LABA, and LABA/LAMA groups were comprised of 58 (32.4%), 52 (29.1%), and 69 (38.5%) patients, respectively. ICS/LABA/LAMA group had a higher severity of bronchiectasis and airflow obstruction, than other groups. In the subgroup with BEC ≥ 300/uL, the risk of moderate-to-severe exacerbation was lower in the ICS/LABA/LAMA group (adjusted HR = 0.137 [95% CI = 0.034–0.553]) and the ICS/LABA group (adjusted HR = 0.196 [95% CI = 0.045–0.861]) compared with the LABA/LAMA group. The annual FEV1 decline rate was significantly worsened in the ICS/LABA group compared to the LABA/LAMA group (adjusted β-coefficient=-197 [95% CI=-307–-87]) in the subgroup with BEC < 200/uL.

Conclusion

In patients with bronchiectasis and airflow obstruction, the use of ICS/LABA/LAMA and ICS/LABA demonstrated a reduced risk of exacerbation compared to LABA/LAMA therapy in those with BEC ≥ 300/uL. Conversely, for those with BEC < 200/uL, the use of ICS/LABA was associated with an accelerated decline in FEV1 in comparison to LABA/LAMA therapy. Further assessment of BEC is necessary as a potential biomarker for the use of ICS in patients with bronchiectasis and airflow obstruction.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12890-024-02867-4.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ABPA
Allergic bronchopulmonary aspergillosis
ACO
Asthma and COPD overlap
BAE
Bronchial artery embolization
BDR
Bronchodilator response
BEC
Blood eosinophil count
BMI
Body mass index
BSI
Bronchiectasis Severity Index
COPD
Chronic obstructive pulmonary disease
CT
Computed tomography
DLCO
Diffusing capacity of the lungs for carbon monoxide
FACED
Forced expiratory volume in 1 s, age, chronic infection with Pseudomonas, radiological extension and dyspnea
FEV1
Forced expiratory volume in 1 s
FVC
Forced vital capacity
GERD
Gastroesophageal reflux disease
hs-CRP
High-sensitivity C-reactive protein
ICS
Inhaled corticosteroid
IQR
Interquartile range
LABA
Long-acting β2-agonist
LAMA
Long-acting muscarinic antagonist
mMRC
Modified Medical Research Council dyspnea scale
NTM–PD
Nontuberculous mycobacteria pulmonary disease
SD
Standard deviation
SE
Standard error
VA
Alveolar volume

Introduction

Bronchiectasis is a chronic airway disease characterised by neutrophilic bronchial inflammation and is commonly reported in the patients with airflow obstruction including asthma [1] or chronic obstructive pulmonary disease (COPD) [2]. As bronchiectasis is diagnosed based on structural abnormality in radiologic evaluation while COPD is diagnosed based on physiologic abnormality in spirometric evaluation, both diagnoses can be fulfilled in a patient with bronchiectasis-COPD overlap (BCO) [3]. With increasing use of screening chest computed tomography (CT) in the patients who ever smoked, BCO has been increasingly documented and the clinical relevance of BCO has been emerging. The patients with bronchiectasis and airflow obstruction had a higher risk of acute exacerbations [4] and mortality [5] than those with bronchiectasis alone. In addition, COPD patients with bronchiectasis had more symptoms, a higher bacterial burden, and a higher risk of acute exacerbation [2].
The effective treatment for bronchiectasis and airflow obstruction has not been sufficiently evaluated. Long-acting beta-2 agonists (LABAs), long-acting muscarinic antagonists (LAMAs), and inhaled corticosteroids (ICSs) have been important drugs for treating COPD. However, their effectiveness in bronchiectasis is less evident [6]. In a randomized controlled trial (RCT), ICS/LABA improved the symptoms and quality of life more than ICS in patients with bronchiectasis [7]. The lung function was numerically more improved with LABA/LAMA than with LABA or LAMA in patients with bronchiectasis [8]. In patients with bronchiectasis, the use of ICS is cautiously considered due to concerns regarding their potential impact on respiratory infections and long-term safety. However, it is still unclear whether inhaled combination therapy with ICS can be beneficial in patients with bronchiectasis and airflow obstruction, especially who had eosinophilia.
The present study aimed to compare the development of acute exacerbation, the change in lung function, and adverse events among patients with bronchiectasis and airflow obstruction treated with ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA.

Materials and methods

The present study followed the guidance presented by the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) statement [9].

Study design and participants

This retrospective study assessed all patients aged ≥ 18 years with forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) < 70% [10] and radiologically confirmed bronchiectasis in chest CT [11] between January 2005 and December 2021 in the Seoul Metropolitan Government–Seoul National University (SMG-SNU) Boramae Medical Center. We included the patients with bronchiectasis and airflow obstruction who underwent baseline and two or more annual follow-up spirometric assessments, experienced an acute exacerbation event during the past year, and were adherent to inhaled combination therapy for at least 6 months. We observed the longest follow-up period during which treatment adherence was appropriate for individual patients. Treatment adherence was assessed by whether inhaled drugs were regularly prescribed. The patients received initial training for the use of the inhaler devices at the first prescription and additional trainings by checking the patient’s technique for the prescribed inhaler devices during follow-up. The eligible patients were classified into 3 groups: ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups.

Pulmonary function test

The highest measured FVC and FEV1 among three or more tests with acceptable curves were used. The absolute values of FVC and FEV1 were obtained, and the percentage of predicted values for FEV1 and FVC were calculated from the Morris equations [12]. Airflow limitation was defined as FEV1/FVC < 0.7 by spirometric evaluation based on the American Thoracic Society/European Respiratory Society guidelines [13]. The positive bronchodilator response (BDR) at baseline was defined as a postbronchodilator increase in FEV1 and/or FVC of at least 12% and 200 mL from baseline values at 15 min after inhalation of 400 µg of salbutamol [14]. Spirometry was conducted by a well-trained technician using a same Vmax series Sensor Medics 2130 automatically computerized spirometry system (SensorMedics) according to official statements of the American Thoracic Society and European Respiratory Society in 2019 [15].

Variables

Baseline information, including age, sex, body mass index (BMI), smoking history, disease severity, previous exacerbation history, bacterial colonization, comorbidities, and treatment duration, was obtained. The history of exacerbations was assessed based on the electronic medical records of the patients. The severity of bronchiectasis was assessed with the Bronchiectasis Severity Index (BSI) and FACED score [16]. Clinical features, including etiology, respiratory symptoms, adjuvant treatments, laboratory tests, spirometric examination, predominant morphology, and number of lobes that were involved, were collected. The basic morphologic types of bronchiectasis (cylindrical, varicose, and cystic) and the involved lobes on chest CT were evaluated by two pulmonologists.

Outcomes

The primary endpoint was to compare the risk of moderate-to-severe exacerbation among the ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups. A moderate exacerbation was defined as an exacerbation leading to treatment with antibiotics or systemic glucocorticoids. A severe exacerbation was one resulting in hospitalization or death [6, 17, 18]. Secondary endpoints were to compare the annual FEV1 change (mL/yr) and the development of adverse events, including pneumonia, MACE, and mortality. The risk of moderate-to-severe exacerbation and the annual FEV1 change were evaluated according to blood eosinophil count (BEC). The baseline measurement of BEC was obtained during the stable phase of the patient’s disease severity.

Statistical analysis

Data are presented as the mean with standard deviation or the median with interquartile range (IQR) for continuous variables and numbers with percentage for categorical variables. Analysis of variance (ANOVA) test was used to test independent samples of continuous, normally distributed data, while the Wilcoxon rank-sum test was used to examine continuous, skewed data. The chi-square test or Fisher’s exact test was used to analyze categorical data. Kaplan–Meier curves and log-rank tests were performed to compare the time to first moderate-to-severe exacerbation among the ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups. We conducted univariable Cox regression analyses for moderate-to-severe exacerbation among the ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups. For multivariable Cox regression analysis, clinically relevant variables were selected through backward elimination method. Clinically relevant variables included age, sex, BMI, current smoking status, mMRC grade, BSI score, FACED score, history of previous moderate-to-severe exacerbation, number of exacerbations in the last 12 months, lung cancer, BEC > 300/uL, high-sensitivity C-reactive protein, baseline FEV1, baseline FEV1/FVC ratio, positive bronchodilator response, chronic infection with Pseudomonas aeruginosa, and radiologic severity. A linear mixed model was used to estimate the effect of the clinical factors contributing to the annual FEV1 change (mL/yr). For multivariable linear mixed model, clinically relevant variables were selected through backward elimination method. P < 0.05 was considered as statistical significance. A variance inflation factor (VIF) > 4.0 was considered as significant multicollinearity. Even though statistical multicollinearity was not confirmed, but high intercorrelation was clinically suspected (e.g. severity score systems and their components), one of the correlated variables was excluded from the multivariable model. We used R statistical software, version 3.6.3 (R Core Team [2020], Vienna, Austria), for statistical analyses.

Ethics

Our study was conducted by following the principles of the Declaration of Helsinki. The institutional review board of the SMG–SNU Boramae Medical Center approved this study and waived the requirement for informed consent (IRB No. 10-2020-099).

Results

Among a total of 355 patients with bronchiectasis and airflow obstruction, 176 patients were excluded because 124 did not undergo at least 2 annual spirometric assessments, 31 were not treated with any inhaled therapy, and 21 were treated with single inhaled therapy. None of the included patients were diagnosed as cystic fibrosis or alpha-1 antitrypsin deficiency. The eligible 179 patients were assigned to the ICS/LABA/LAMA group (n = 58), the ICS/LABA group (n = 52), and the LABA/LAMA group (n = 69) (Fig. 1). They underwent a median of 4 (IQR = 3–5) annual spirometric assessments, and their median annual FEV1 change was − 89 (IQR= -364–291) mL/yr. The median follow-up duration was 40 [IQR = 23–62] months.

Baseline characteristics and clinical features

The baseline characteristics of the included patients are described in Table 1. There were significant differences in sex, BMI, smoking history, disease severity, and previous exacerbation history between the three groups. The ICS/LABA/LAMA group showed a lower BMI, a higher likelihood of smoking, a greater severity of bronchiectasis, and a history of more severe previous exacerbation event compared to both the LABA/LAMA and ICS/LABA groups. Considering the FACED score, the LABA/LAMA group had a higher severity of bronchiectasis than the ICS/LABA group. The proportion of overall detected bacteria that colonized the lungs was 31% and was comparable between the three groups. The treatment duration with inhaled combination therapy was significantly longer in the ICS/LABA/LAMA and ICS/LABA groups than in the LABA/LAMA group. The results of post-hoc analysis are described in Supplementary information 1 and 2.
Table 1
Baseline characteristics of patients with bronchiectasis and airflow obstruction treated with inhaled combination therapies
 
ICS/LABA/LAMA (n = 58)
ICS/LABA
(n = 52)
LABA/LAMA (n = 69)
P-value
Age, mean (SD)
61.05 (10.35)
62.60 (12.19)
63.81 (11.34)
0.392
Female, n (%)
7 (12.1)
20 (38.5)
15 (21.7)
0.004
BMI, mean (SD)
20.44 (4.06)
22.86 (4.42)
21.86 (3.66)
0.011
Smoking history
    
Never smoker, n (%)
10 (17.2)
20 (38.5)
25 (36.2)
0.025
Ex-smoker, n (%)
25 (43.1)
18 (34.6)
29 (42.0)
0.615
Current smoker, n (%)
23 (39.7)
14 (26.9)
15 (21.7)
0.079
Pack years, median (IQR)
30 (22–38)
15 (9–21)
20 (14–26)
0.002
Disease severity
    
mMRC score, mean (SD)
2.09 (0.82)
1.63 (0.66)
1.75 (0.81)
0.007
BSI score, mean (SD)
7.91 (3.75)
5.29 (2.80)
6.06 (3.25)
< 0.001
FACED score, mean (SD)
2.66 (1.66)
1.63 (1.69)
2.29 (1.48)
0.004
Previous moderate or severe exacerbation history, n (%)
24 (43.6)
14 (29.2)
14 (20.3)
0.019
Bacterial colonizer, n (%)
21 (36.2)
14 (26.9)
21 (30.4)
0.566
Pseudomonas colonizer, n (%)
7 (12.1)
2 (3.8)
6 (9.2)
0.298
Comorbidity
    
Hypertension, n (%)
24 (41.4)
24 (46.2)
33 (47.8)
0.759
Diabetes mellitus, n (%)
11 (19.0)
17 (32.7)
20 (29.0)
0.234
Chronic kidney disease, n (%)
6 (10.3)
3 (5.8)
6 (8.7)
0.683
Chronic liver disease, n (%)
6 (10.3)
7 (13.5)
10 (14.5)
0.775
Cerebrovascular disease, n (%)
6 (10.3)
6 (11.5)
7 (10.1)
0.967
Cardiovascular disease, n (%)
9 (15.5)
13 (25.0)
14 (20.3)
0.464
Lung cancer, n (%)
7 (12.1)
4 (7.7)
15 (21.7)
0.077
Malignancy other than lung cancer, n (%)
10 (17.2)
12 (23.1)
9 (13.0)
0.353
Duration of inhaled combination therapy, month, mean (SD)
62.81 (39.15)
55.96 (44.83)
33.34 (17.63)
< 0.001
Note: Data presented as n (%) for categorical variables or mean (SD) or median (IQR) for numerical variables
Abbreviations: ICS, inhaled corticosteroid; LABA, long-acting β2-agonist; LAMA, long-acting muscarinic antagonist; BMI, body mass index; mMRC, Modified Medical Research Council dyspnea scale; BSI, Bronchiectasis Severity Index; FACED, forced expiratory volume in 1 s, age, chronic infection with Pseudomonas, radiological extension and dyspnea; COPD, chronic obstructive pulmonary disease; ACO, Asthma and COPD overlap; NTM?PD, nontuberculous mycobacteria pulmonary disease
Regarding the clinical features, we found no difference in the etiology of bronchiectasis among the three groups (Table 2). Non-purulent sputum was more abundant, and mucolytics, including N-acetylcysteine and erdosteine, were more commonly used in the ICS/LABA/LAMA group than in the LABA/LAMA and ICS/LABA groups. In addition, the proportion of patients requiring long–term oxygen therapy was significantly higher in the ICS/LABA/LAMA group. ICS/LABA/LAMA and ICS/LABA groups showed a higher BEC compared to LABA/LAMA group. At the baseline spirometric examination, FEV1 (%) and FEV1/FVC (%) were significantly lower in the ICS/LABA/LAMA and LABA/LAMA groups than in the ICS/LABA group. The ICS/LABA group had a higher FVC (%) than the LABA/LAMA group and a higher DLCO/VA (%) than the ICS/LABA/LAMA group. The BDR positivity was significantly lower in LABA/LAMA than in ICS/LABA and ICS/LABA/LAMA. There was no difference in the morphologic features or the involved pulmonary lobes of bronchiectasis in chest CT among the three groups.
Table 2
Clinical features of patients with bronchiectasis and airflow obstruction treated with inhaled combination therapies
 
ICS/LABA/LAMA
(n = 58)
ICS/LABA
(n = 52)
LABA/LAMA
(n = 69)
P-value
Etiology of bronchiectasis
    
Post-infectious, n (%)
28 (48.3)
14 (26.9)
29 (42.0)
0.064
Idiopathic, n (%)
16 (27.6)
18 (34.6)
24 (34.8)
0.635
Chronic airway disease, n (%)
9 (15.5)
10 (19.2)
8 (11.6)
0.506
ABPA, n (%)
2 (3.4)
5 (9.6)
3 (4.3)
0.316
GERD, n (%)
2 (3.4)
4 (7.7)
3 (4.3)
0.565
Connective tissue disease, n (%)
0
1 (1.9)
1 (1.4)
0.598
Immunosuppression, n (%)
1 (1.7)
0 (0.0)
1 (1.4)
0.654
Respiratory symptoms
    
Cough, n (%)
25 (43.1)
23 (44.2)
25 (36.2)
0.614
Non-purulent sputum, n (%)
20 (34.5)
8 (15.4)
7 (10.1)
0.002
Purulent sputum, n (%)
12 (20.7)
9 (17.3)
17 (24.6)
0.616
Hemoptysis, n (%)
11 (19.0)
9 (17.3)
18 (26.1)
0.443
Chest discomfort, n (%)
2 (3.4)
1 (1.9)
1 (1.4)
0.738
Dyspnea, n (%)
45 (77.6)
34 (65.4)
45 (65.2)
0.248
Adjuvant treatments
    
N-acetylcystein, n (%)
19 (32.8)
9 (17.3)
11 (15.9)
0.048
Ambroxol, n (%)
19 (32.8)
11 (21.2)
20 (29.0)
0.388
Erdosteine, n (%)
39 (67.2)
26 (50.0)
31 (44.9)
0.035
Bronchial artery embolization history, n (%)
9 (15.5)
4 (7.7)
14 (20.3)
0.158
Long–term oxygen therapy, n (%)
47 (81.0)
33 (64.7)
41 (59.4)
0.029
Laboratory tests
    
White blood cell, 1000/uL mean (SD)
9.54 (13.03)
7.65 (2.39)
8.03 (2.87)
0.391
Hemoglobin, g/dl, mean (SD)
14.45 (3.69)
13.46 (1.48)
13.02 (1.74)
0.006
Platelet, 1000/uL, mean (SD)
248 (79)
266 (87)
248 (93)
0.459
Blood eosinophil count, /uL, mean (SD)
408 (223)
467 (355)
239 (223)
< 0.001
Blood eosinophil count, n (%)
    
< 150/uL
0
0
28 (41.2)
< 0.001
150–299/uL
20 (34.5)
22 (42.3)
23 (33.8)
0.529
≥ 300/uL
38 (65.5)
30 (57.7)
17 (25.0)
< 0.001
hs-CRP, median (IQR)
0.78 (0–2.05)
0.35 (0–0.70)
1.56 (0.89–2.23)
0.001
Spirometric examination
    
FVC, L, mean (SD)
2.79 (0.72)
2.63 (0.98)
2.43 (0.69)
0.035
FVC, %, mean (SD)
76.38 (17.34)
79.75 (20.02)
70.80 (16.84)
0.023
FEV1, L, mean (SD)
1.22 (0.43)
1.44 (0.61)
1.19 (0.44)
0.016
FEV1, %, mean (SD)
47.33 (15.07)
62.87 (22.85)
49.86 (15.56)
< 0.001
FEV1/FVC, %, mean (SD)
44.13 (12.32)
56.02 (13.37)
49.66 (12.96)
< 0.001
DLCO, L, mean (SD)
10.73 (4.88)
13.10 (5.59)
10.89 (5.16)
0.046
DLCO, %, mean (SD)
63.54 (22.29)
75.23 (27.99)
66.89 (27.90)
0.083
DLCO/VA, L, mean (SD)
2.92 (1.13)
3.54 (1.15)
3.08 (1.26)
0.031
DLCO/VA, %, mean (SD)
76.85 (27.87)
93.25 (28.10)
82.11 (31.59)
0.022
BDR positivity, n (%)
15 (25.9)
16 (30.8)
6 (8.8)
0.007
Predominant morphology in CT
    
Cylindrical, n (%)
27 (46.6)
22 (42.3)
22 (31.9)
0.218
Varicose, n (%)
17 (29.3)
18 (34.6)
29 (42.0)
0.323
Cystic, n (%)
14 (24.1)
12 (23.1)
18 (26.1)
0.926
Total number of lobe involvement in CT, mean (SD)
2.64 (1.53)
2.29 (1.42)
2.75 (1.58)
0.238
Note: Data presented as n (%) for categorical variables or mean (SD) or median (IQR) for numerical variables
Abbreviations: ABPA, allergic bronchopulmonary aspergillosis; BAE, bronchial artery embolization; BDR, bronchodilator response; CT, computed tomography; DLCO, diffusing capacity of the lungs for carbon monoxide; FVC, forced vital capacity; FEV1, forced expiratory volume in 1 second; GERD, gastroesophageal reflux disease; ICS, inhaled corticosteroid; LABA, long-acting β2-agonist; LAMA, long-acting muscarinic antagonist; SD, standard deviation; SE, standard error; VA, alveolar volume

Exacerbation

The number of moderate-to-severe exacerbation events was not significantly different among the three groups. However, the time to the first event of a moderate-to-severe exacerbation was significantly shorter in the LABA/LAMA group than in the ICS/LABA and ICS/LABA/LAMA groups (log-rank test, P-value < 0.001, Fig. 2). In the univariable Cox regression model, older age, a higher grade of mMRC, a higher score of BSI, a higher score of FACED, previous history of moderate-to-severe exacerbation, and a higher number of exacerbations in previous year were related to an increased hazard of a moderate–to–severe exacerbation in the patients with bronchiectasis and airflow obstruction (Table 3). However, in the multivariable Cox regression analysis, the hazard of moderate-to-severe exacerbation in the ICS/LABA and ICS/LABA/LAMA groups was not significantly different from the LABA/LAMA groups (ICS/LABA vs. LABA/LAMA, adjusted HR = 0.491 [95% CI = 0.191–1.263], P-value = 0.140; ICS/LABA/LAMA vs. LABA/LAMA, adjusted HR = 0.692 [95% CI = 0.293–1.638], P-value = 0.403) The adjusted hazard for moderate-to-severe exacerbation was not statistically different between the ICS/LABA and ICS/LABA/LAMA groups.
Table 3
Hazard ratio of moderate-to-severe exacerbation in patients with bronchiectasis and airflow obstruction treated with inhaled combination therapies
 
Univariable Cox regression model
Multivariable Cox regression model
HR (95% CI)
P-value
HR (95% CI)
P-value
Age
1.030 (1.009–1.052)
0.006
1.035 (1.012–1.059)
0.003
Female
0.898 (0.563–1.430)
0.649
  
BMI
1.019 (0.968–1.072)
0.476
  
Current smoker
1.276 (0.812–2.007)
0.291
2.041 (1.209–3.447)
0.007
mMRC grade
1.408 (1.081–1.834)
0.011
1.315 (1.012–1.7309)
0.030
BSI score
1.076 (1.019–1.137)
0.009
  
FACED score
1.140 (1.015–1.280)
0.027
  
Previous moderate or severe exacerbation history
1.546 (1.005–2.379)
0.047
  
Number of exacerbations in previous year
2.539 (2.002–3.220)
< 0.001
1.617 (1.017–2.571)
< 0.037
Lung cancer
1.309 (0.705–2.430)
0.394
  
Blood eosinophil count > 300/uL
0.559 (0.369–0.845)
0.006
0.583 (0.350–0.971)
0.038
hs-CRP
1.001 (0.943–1.064)
0.962
  
Baseline FEV1(L)
0.614 (0.364–1.037)
0.068
  
Baseline FEV1/FVC (%)
0.997 (0.981–1.013)
0.703
  
Baseline DLCO, (%)
0.852 (0.696–1.043)
0.120
0.988 (0.977–0.998)
0.024
Positive bronchodilator response
0.767 (0.475–1.238)
0.277
  
Colonization with Pseudomonasaeruginosa
1.710 (0.746–3.918)
0.205
  
Total number of lobe involvement in CT
0.981 (0.851–1.130)
0.788
  
Inhaled combination therapy (ref: LABA/LAMA)
   
ICS/LABA
0.369 (0.207–0.658)
< 0.001
0.491 (0.191–1.263)
0.140
ICS/LABA/LAMA
0.413 (0.244–0.699)
< 0.001
0.692 (0.293–1.638)
0.403
Note: Data were analyzed with univariable and multivariable cox regression models and are presented as adjusted hazard ratio (95% confidence interval). BSI and FACED were omitted from the multivariable analysis due to concerns of collinearity with other clinical variables.
Abbreviations: BMI, body mass index; BSI, Bronchiectasis Severity Index; CI, confidence interval; COPD, chronic obstructive pulmonary disease; CT, computed tomography; DLCO, diffusing capacity of the lungs for carbon monoxide; FACED, forced expiratory volume in 1 s, age, chronic infection with Pseudomonas, radiological extension and dyspnea; FEV1, forced expiratory volume in 1 second; FVC, forced vital capacity; HR, hazard ratio; hs-CRP, high-sensitivity C-reactive protein; ICS, inhaled corticosteroid; LABA, long-acting β2-agonist; LAMA, long-acting muscarinic antagonist; mMRC, Modified Medical Research Council dyspnea scale

Lung function decline rate

In the multivariable linear mixed effect model, elderly, female, a lower BMI, current smoker, a higher grade of mMRC, a lower baseline FEV1, and previous history of moderate-to-severe exacerbation were related with accelerated annual FEV1 decline rate (Table 4). There was no significant difference in annual FEV1 decline rate among the ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups.
Table 4
Effect of clinical factors contributing to the annual FEV1 change (mL/yr)
 
Adjusted β-coefficient (95% CI)
P-value
Age
-4.18 (-6.19–-2.17)
< 0.001
Female
-121.15 (-172.84–-69.47)
< 0.001
BMI
11.29 (5.99–16.59)
< 0.001
Current smoker
-76.61 (-123.54–-29.69)
0.001
mMRC grade
-34.12 (-62.31–-5.93)
0.018
Baseline FEV1(100 mL)
75.25 (70.17–80.32)
< 0.001
Previous moderate or severe exacerbation history
-50.51 (-97.51–-3.50)
0.036
Inhaled therapy (ref: LABA/LAMA)
  
ICS/LABA
-49.73 (-105.47–6.01)
0.081
ICS/LABA/LAMA
48.83 (-1.94–99.59)
0.060
Note: Data were analyzed with mixed linear regression and are presented as linear regression coefficient and standard error. BSI and FACED were omitted from the multivariable analysis due to concerns of collinearity with other clinical variables.
Abbreviations: BMI, body mass index; FEV1, forced expiratory volume in 1 second; LABA, long-acting β2-agonist; LAMA, long-acting muscarinic antagonist

Subgroup analysis according to BEC

The ICS/LABA/LAMA group had a lower risk of moderate-to-severe exacerbation in subgroup with BEC ≥ 300/uL (adjusted HR = 0.137 [95% CI = 0.034–0.553], P-value = 0.005) than LABA/LAMA group (Table 5). In the subgroup with BEC ≥ 300/uL, annual FEV1 decline rate was numerically more attenuated without statistical significance in the ICS/LABA/LAMA group compared to LABA/LAMA group (adjusted β-coefficient = 246.45 [95% CI=-63.80–556.70]], P-value = 0.128) and in the subgroup with BEC = 150–299/uL (adjusted β-coefficient = 191.80 [95% CI=-39.03–422.64], P-value = 0.123).
Table 5
Acute exacerbation and annual FEV1 change according to the blood eosinophil count
 
Moderate-to-severe exacerbation
Annual FEV1 change, mL/yr
 
adjusted HRa
(95% CI)
P-value
Adjusted β-coefficientb
(95% CI)
P-value
Blood eosinophil count, ≥ 300/uL
    
ICS/LABA (compared with LABA/LAMA)
0.196 (0.045–0.861)
0.031
-137.06 (-428.66–154.53)
0.361
ICS/LABA/LAMA (compared with LABA/LAMA)
0.137 (0.034–0.553)
0.005
246.45 (-63.80–556.70)
0.128
Blood eosinophil count, 200–299/uL
    
ICS/LABA (compared with LABA/LAMA)
0.944 (0.175–5.101)
0.947
-42.72 (-398.09–312.65)
0.817
ICS/LABA/LAMA (compared with LABA/LAMA)
0.655 (0.098–4.388)
0.663
191.80 (-39.03–422.64)
0.123
Blood eosinophil count, < 200/uL
    
ICS/LABA (compared with LABA/LAMA)
1.918 (0.465–7.908)
0.368
-197.18 (-307.04–-87.32)
< 0.001
ICS/LABA/LAMA (compared with LABA/LAMA)
1.467 (0.462–4.658)
0.515
9.63 (-115.23–134.49)
0.880
Note: Data were analyzed with mixed linear regression and are presented as linear regression coefficient and standard error
a Adjusted hazard ratio was estimated using covariables including age, current smoker, mMRC grade, baseline FEV1, and number of exacerbations in previous year.
b Adjusted β-coefficient was estimated using covariables including age, sex, BMI, current smoker, mMRC grade, baseline FEV1, and previous history of moderate or severe exacerbation.
ICS/LABA group showed a lower risk of moderate-to-severe exacerbation (adjusted HR = 0.196 [95% CI = 0.045–0.861], P-value = 0.005) compared to LABA/LAMA group in the subgroup with BEC ≥ 300/uL (Table 5). In addition, annual FEV1 decline rate was more accelerated in the ICS/LABA group compared to LABA/LAMA group in the subgroup with BEC < 200/uL (adjusted β-coefficient=-197.18 [95% CI=-307.04–-87.32], P-value < 0.001).

Adverse events

There was no significant difference in the development of pneumonia between the ICS/LABA/LAMA (n = 40, 69.0%), ICS/LABA (n = 32, 61.5%), and LABA/LAMA (n = 44, 62.9%) groups. MACE was similarly reported among the ICS/LABA/LAMA (n = 8, 13.8%), ICS/LABA (n = 11, 21.2%), and LABA/LAMA (n = 14, 20.0%) groups. We found no difference in mortality events among the ICS/LABA/LAMA (n = 7, 12.1%), ICS/LABA (n = 2, 3.8%), and LABA/LAMA (n = 7, 10.0%) groups.

Discussion

Our longitudinal observational study compared the efficacy and safety of ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA treatments in the patients with bronchiectasis and airflow obstruction. In the baseline clinical features, more symptoms, greater severity of bronchiectasis, and a history of more frequent exacerbations were found in the ICS/LABA/LAMA group than in the LABA/LAMA or ICS/LABA groups. In addition, the ICS/LABA/LAMA group had a lower baseline FEV1 and FEV1/FVC than the ICS/LABA group. Despite these differences, the ICS/LABA/LAMA group did not show a significant difference in the adjusted HR for moderate-to-severe exacerbation compared to the ICS/LABA group and LABA/LAMA group. However, the beneficial effect of ICS/LABA/LAMA and ICS/LABA in reducing moderate-to-severe exacerbation was observed in patients with BEC ≥ 300/uL. There was no significant difference in reducing the annual FEV1 decline rate among the ICS/LABA/LAMA, ICS/LABA, and LABA/LAMA groups. However, ICS/LABA was associated with an accelerated FEV1 decline in those with BEC ≥ 300/uL compared to the LABA/LAMA group. We found no difference in the incidence of pneumonia, MACE, or death among the three groups. Combination therapy with ICS may have benefits in preventing clinical deterioration in the patients with bronchiectasis and airway obstruction in the presence of high BEC.
There have been several efforts to determine the beneficial effects of inhaled therapies in bronchiectasis. In a prospective study with 77 patients, budesonide group showed numerically less exacerbations and more improvement of FEV1 without statistical significance [19]. Another prospective study analyzed the effect of inhaled beclomethasone diproprionate for 6 weeks in 20 patients with bronchiectasis and revealed a significant improvement in the FEV1 [20]. In a clinical trial, inhaled combination therapy with medium-dose budesonide and formoterol was compared for a year with high-dose budesonide in patients with bronchiectasis [7]. In this study, patients with medium-dose budesonide and formoterol had a better improvement in symptoms but did not show improvements in their lung function or in reducing acute exacerbation [7]. However, those results from previous studies have been interpreted limitedly in that the number of study participants was small (fewer than 100) and the treatment duration was as short as a year or less. Compared with previous studies, we included the patients with bronchiectasis who had airflow limitation (FEV1/FVC < 0.7) and followed them up for a longer period (more than 3 years). We could find a potential role of ICS for the patients with eosinophilic bronchiectasis and airflow obstruction, who had BEC ≥ 300/uL. More studies are needed to clarify the potential benefits of using inhaled combination therapy with ICS among patients with eosinophilic bronchiectasis and airflow obstruction.
In our study, the ICS/LABA/LAMA and ICS/LABA groups were associated with a lower exacerbation risk than the LABA/LAMA group when the bacterial load including Pseudomonas aeruginosa (PA) was 31% and comparable between the three groups. The prevalence of PA infection or colonizer in non-cystic bronchiectasis has been reported from 9 to 34% in several studies [2228]. In individuals diagnosed with non-cystic fibrosis bronchiectasis, infection with PA is correlated with heightened sputum production, diminished lung function, and a deceleration of respiratory ciliary beat in vivo [24, 27, 29]. Interestingly, a retrospective study showed a reduction in the exacerbation frequency in patients with PA infection who were treated with inhaled fluticasone propionate [21]. The interaction between PA and the respiratory mucosa remains inadequately elucidated, with the role of corticosteroids in this process presenting further ambiguity. The observed effectiveness of ICS treatment within this specific patient subgroup implies a potential beneficial role of the interaction between PA or its toxins and the bronchiectatic airways. Given the absence of established treatments for chronic PA infection in the airways of these patients, there is an imperative need to scrutinize the underlying mechanism(s) driving this phenomenon.
Although plausible mechanisms for the beneficial role of ICS in bronchiectasis and airflow obstruction have not been well explored, ICS reportedly suppresses airway inflammation in selected patients with bronchiectasis [30] and COPD [31]. ICS reduced the sputum production and decreased the levels of leucocytes, IL–1b, IL–8, and LTB4 in the sputum [32]. The improvement in the sputum volume is assumed to be the consequence of the downregulation of airway proinflammatory mediators. Decreased inflammatory mediators by ICS could lead to amelioration of leucocyte trafficking, less neutrophilic infiltration, and less release of toxic products into the bronchiectatic airway [33]. In a clinical trial, high-dose ICS reduced the sputum production and improved the symptoms in patients with bronchiectasis [34]. In addition, the complementary mechanism of ICS and LABA may affect the clinical outcomes in bronchiectasis patients. The anti-inflammatory effect of ICS was greater with the concurrent use of beta-agonists through an enhanced translocation of the glucocorticoid receptor or through the potentiated molecular mechanisms of glucocorticoids [35, 36]. In addition, ICS also increased the number of beta-2 receptors or prevented the downregulation of beta-2 receptors by activating gene transcription [3638]. The synergistic anti-inflammatory effect of ICS/LABAs may overweigh the enhanced bronchodilating effect of LABAs/LAMAs in patients with bronchiectasis and airflow obstruction. Currently, BEC has been considered as a biomarker to identify the subgroup COPD patients who can benefit from ICS treatment. Several post-hoc analyses of clinical trials, utilizing different thresholds for BEC have reported a better response to ICS in patients with a higher baseline BEC [39]. Recent prospective studies have reported a better ICS response for reducing exacerbation in the patients with a higher BEC [40, 41]. Our study suggests that eosinophil can be an important biomarker to predict the response to ICS in the patients with bronchiectasis and airflow obstruction.
There is a paucity of data on the association between inhaled bronchodilators and the clinical course of patients with bronchiectasis and airflow obstruction. Lung function was more improved when bronchiectasis patients were treated with inhaled bronchodilators, especially in patients with a positive bronchodilator response [8, 42, 43]. Adding formoterol to the ICS therapy was related to improved symptoms in the patients with bronchiectasis [7]. A recent RCT showed that tiotropium improved lung function over 6 months in stable patients with bronchiectasis who showed airflow limitations (44). However, benefits in reducing exacerbations or mortality by LABA or LAMA have not been reported in patients with bronchiectasis. In the present study, there were no significant differences in acute exacerbation and lung function decline rate between the ICS/LABA and ICS/LABA/LAMA groups. Considering the greater baseline disease severity of the ICS/LABA/LAMA group compared with ICS/LABA group, there may be a beneficial role of LAMA in patients with bronchiectasis and airflow obstruction.
This study has several limitations. First, our retrospective study analyzed a small number of patients with bronchiectasis and airflow obstruction who used inhaled combination therapy. As ICS tends to be underused in bronchiectasis, our patients are considered to represent a distinct subpopulation of patients with bronchiectasis. Therefore, our results cannot be generalizable to all patients with bronchiectasis. Furthermore, it was challenging to figure out the actual adherence rates or adequate technique rates for inhalers due to the nature of retrospective assessment. More studies with a larger number of patients are needed to generalize the potential benefits of inhaled combination therapy in bronchiectasis and airflow obstruction. Second, it was difficult to distinguish whether the benefit of ICS was related with pathogenesis of bronchiectasis. However, it was found that the benefit of ICS outweighs the potential harm to bronchiectasis in patients with airflow obstruction and eosinophilia. The effect of ICS on eosinophilic inflammation may benefit beyond COPD to bronchiectasis. Third, it was still questionable whether inhaled combination therapy has better clinical outcomes than single inhaled therapy. Because of large clinical heterogeneities, comparisons between single inhaled therapy and inhaled combination therapy cannot be performed properly in retrospective study designs. Instead, we limitedly assumed the additional benefit of ICS while comparing ICS/LABA/LAMA and LABA/LAMA and the additional benefit of LAMA while comparing ICS/LABA/LAMA and ICS/LABA.

Conclusion

ICS/LABA/LAMA or ICS/LABA may be related with a lower risk of acute exacerbation compared with LABA/LAMA in patients with bronchiectasis and airflow obstruction, especially who had a higher BEC. The annual FEV1 decline rate was significantly worsened in the ICS/LABA group compared to the LABA/LAMA group in those with BEC < 200/uL. BEC needs to be further evaluated as a biomarker before the use of ICS in the patients with bronchiectasis and airflow obstruction.

Acknowledgements

Not applicable.

Competing interests

The authors declare no competing interests.

Conflict of interest disclosure

All authors declare no conflicts of interest for the present study.

Ethics approval statement

Our study was conducted in accordance with the ethical guidance of the Declaration of Helsinki in 1975. The Institutional Review Board Committee of Seoul National University Seoul Metropolitan Government (SNU-SMG) Boramae Medical Center approved the study protocol and waived the requirement for informed consent from study subjects for access to the electronic medical records (IRB No. 10-2020-099).
Not applicable.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Oguzulgen IK, Kervan F, Ozis T, Turktas H. The impact of bronchiectasis in clinical presentation of asthma. South Med J. 2007;100(5):468–71.CrossRefPubMed Oguzulgen IK, Kervan F, Ozis T, Turktas H. The impact of bronchiectasis in clinical presentation of asthma. South Med J. 2007;100(5):468–71.CrossRefPubMed
2.
3.
Zurück zum Zitat Hurst JR, Elborn JS, Soyza AD. COPD–bronchiectasis overlap syndrome. Eur Respir J. 2015;45(2):310–3.CrossRefPubMed Hurst JR, Elborn JS, Soyza AD. COPD–bronchiectasis overlap syndrome. Eur Respir J. 2015;45(2):310–3.CrossRefPubMed
4.
Zurück zum Zitat Patel IS, Vlahos I, Wilkinson TM, Lloyd-Owen SJ, Donaldson GC, Wilks M, et al. Bronchiectasis, exacerbation indices, and inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2004;170(4):400–7.CrossRefPubMed Patel IS, Vlahos I, Wilkinson TM, Lloyd-Owen SJ, Donaldson GC, Wilks M, et al. Bronchiectasis, exacerbation indices, and inflammation in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2004;170(4):400–7.CrossRefPubMed
5.
Zurück zum Zitat Martínez-García MA, de la Rosa Carrillo D, Soler-Cataluña JJ, Donat-Sanz Y, Serra PC, Lerma MA, et al. Prognostic value of bronchiectasis in patients with moderate-to-severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;187(8):823–31.CrossRefPubMed Martínez-García MA, de la Rosa Carrillo D, Soler-Cataluña JJ, Donat-Sanz Y, Serra PC, Lerma MA, et al. Prognostic value of bronchiectasis in patients with moderate-to-severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2013;187(8):823–31.CrossRefPubMed
6.
Zurück zum Zitat Pasteur MC, Bilton D, Hill AT. British thoracic society guideline for non-CF bronchiectasis. Thorax. 2010;65(Suppl 1):i1–58.CrossRefPubMed Pasteur MC, Bilton D, Hill AT. British thoracic society guideline for non-CF bronchiectasis. Thorax. 2010;65(Suppl 1):i1–58.CrossRefPubMed
7.
Zurück zum Zitat Martínez-García M, Soler-Cataluña JJ, Catalán-Serra P, Román-Sánchez P, Tordera MP. Clinical efficacy and safety of budesonide-formoterol in non-cystic fibrosis bronchiectasis. Chest. 2012;141(2):461–8.CrossRefPubMed Martínez-García M, Soler-Cataluña JJ, Catalán-Serra P, Román-Sánchez P, Tordera MP. Clinical efficacy and safety of budesonide-formoterol in non-cystic fibrosis bronchiectasis. Chest. 2012;141(2):461–8.CrossRefPubMed
8.
Zurück zum Zitat Lee SY, Lee JS, Lee SW, Oh YM. Effects of treatment with long-acting muscarinic antagonists (LAMA) and long-acting beta-agonists (LABA) on lung function improvement in patients with bronchiectasis: an observational study. J Thorac Disease. 2021;13(1):169–77.CrossRef Lee SY, Lee JS, Lee SW, Oh YM. Effects of treatment with long-acting muscarinic antagonists (LAMA) and long-acting beta-agonists (LABA) on lung function improvement in patients with bronchiectasis: an observational study. J Thorac Disease. 2021;13(1):169–77.CrossRef
9.
Zurück zum Zitat von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP. The strengthening the reporting of Observational studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Lancet. 2007;370(9596):1453–7.CrossRef von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC, Vandenbroucke JP. The strengthening the reporting of Observational studies in Epidemiology (STROBE) statement: guidelines for reporting observational studies. Lancet. 2007;370(9596):1453–7.CrossRef
10.
Zurück zum Zitat 2022 Global Strategy for diagnosis, Management and Prevention of chronic obstructive pulmonary disease. Global Initiative for Chronic Obstructive Lung Disease. 2022 Global Strategy for diagnosis, Management and Prevention of chronic obstructive pulmonary disease. Global Initiative for Chronic Obstructive Lung Disease.
11.
Zurück zum Zitat Díaz AA, Nardelli P, Wang W, San José Estépar R, Yen A, Kligerman S et al. Artificial Intelligence-based CT Assessment of Bronchiectasis: the COPDGene Study. Radiology. 2022:221109. Díaz AA, Nardelli P, Wang W, San José Estépar R, Yen A, Kligerman S et al. Artificial Intelligence-based CT Assessment of Bronchiectasis: the COPDGene Study. Radiology. 2022:221109.
12.
Zurück zum Zitat Morris JF, Koski A, Johnson LC. Spirometric standards for healthy nonsmoking adults. Am Rev Respir Dis. 1971;103(1):57–67.PubMed Morris JF, Koski A, Johnson LC. Spirometric standards for healthy nonsmoking adults. Am Rev Respir Dis. 1971;103(1):57–67.PubMed
13.
Zurück zum Zitat Quanjer PH, Pretto JJ, Brazzale DJ, Boros PW. Grading the severity of airways obstruction: new wine in new bottles. Eur Respir J. 2014;43(2):505–12.CrossRefPubMed Quanjer PH, Pretto JJ, Brazzale DJ, Boros PW. Grading the severity of airways obstruction: new wine in new bottles. Eur Respir J. 2014;43(2):505–12.CrossRefPubMed
14.
Zurück zum Zitat Pellegrino R, Viegi G, Brusasco V, Crapo RO, Burgos F, Casaburi R, et al. Interpretative strategies for lung function tests. Eur Respir J. 2005;26(5):948–68.CrossRefPubMed Pellegrino R, Viegi G, Brusasco V, Crapo RO, Burgos F, Casaburi R, et al. Interpretative strategies for lung function tests. Eur Respir J. 2005;26(5):948–68.CrossRefPubMed
15.
Zurück zum Zitat Graham BL, Steenbruggen I, Miller MR, Barjaktarevic IZ, Cooper BG, Hall GL, et al. Standardization of Spirometry 2019 Update. An official American Thoracic Society and European Respiratory Society Technical Statement. Am J Respir Crit Care Med. 2019;200(8):e70–e88.CrossRefPubMedPubMedCentral Graham BL, Steenbruggen I, Miller MR, Barjaktarevic IZ, Cooper BG, Hall GL, et al. Standardization of Spirometry 2019 Update. An official American Thoracic Society and European Respiratory Society Technical Statement. Am J Respir Crit Care Med. 2019;200(8):e70–e88.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Costa JC, Machado JN, Ferreira C, Gama J, Rodrigues C. The Bronchiectasis Severity Index and FACED score for assessment of the severity of bronchiectasis. Pulmonology. 2018. Costa JC, Machado JN, Ferreira C, Gama J, Rodrigues C. The Bronchiectasis Severity Index and FACED score for assessment of the severity of bronchiectasis. Pulmonology. 2018.
17.
Zurück zum Zitat Lipson DA, Barnhart F, Brealey N, Brooks J, Criner GJ, Day NC, et al. Once-daily single-inhaler Triple versus Dual Therapy in patients with COPD. N Engl J Med. 2018;378(18):1671–80.CrossRefPubMed Lipson DA, Barnhart F, Brealey N, Brooks J, Criner GJ, Day NC, et al. Once-daily single-inhaler Triple versus Dual Therapy in patients with COPD. N Engl J Med. 2018;378(18):1671–80.CrossRefPubMed
18.
Zurück zum Zitat Agustí A, Celli BR, Criner GJ, Halpin D, Anzueto A, Barnes P, et al. Global Initiative for Chronic Obstructive Lung Disease 2023 Report: GOLD Executive Summary. Am J Respir Crit Care Med. 2023;207(7):819–37. Agustí A, Celli BR, Criner GJ, Halpin D, Anzueto A, Barnes P, et al. Global Initiative for Chronic Obstructive Lung Disease 2023 Report: GOLD Executive Summary. Am J Respir Crit Care Med. 2023;207(7):819–37.
19.
Zurück zum Zitat Hernando R, Drobnic ME, Cruz MJ, Ferrer A, Suñé P, Montoro JB, et al. Budesonide efficacy and safety in patients with bronchiectasis not due to cystic fibrosis. Int J Clin Pharm. 2012;34(4):644–50.CrossRefPubMed Hernando R, Drobnic ME, Cruz MJ, Ferrer A, Suñé P, Montoro JB, et al. Budesonide efficacy and safety in patients with bronchiectasis not due to cystic fibrosis. Int J Clin Pharm. 2012;34(4):644–50.CrossRefPubMed
20.
Zurück zum Zitat Elborn JS, Johnston B, Allen F, Clarke J, McGarry J, Varghese G. Inhaled steroids in patients with bronchiectasis. Respir Med. 1992;86(2):121–4.CrossRefPubMed Elborn JS, Johnston B, Allen F, Clarke J, McGarry J, Varghese G. Inhaled steroids in patients with bronchiectasis. Respir Med. 1992;86(2):121–4.CrossRefPubMed
21.
22.
Zurück zum Zitat Loebinger MR, Wells AU, Hansell DM, Chinyanganya N, Devaraj A, Meister M, et al. Mortality in bronchiectasis: a long-term study assessing the factors influencing survival. Eur Respir J. 2009;34(4):843–9.CrossRefPubMed Loebinger MR, Wells AU, Hansell DM, Chinyanganya N, Devaraj A, Meister M, et al. Mortality in bronchiectasis: a long-term study assessing the factors influencing survival. Eur Respir J. 2009;34(4):843–9.CrossRefPubMed
23.
Zurück zum Zitat PASTEUR MC, HELLIWELL SM, HOUGHTON SJ, WEBB SC, FOWERAKER JE, COULDEN RA, et al. An investigation into causative factors in patients with Bronchiectasis. Am J Respir Crit Care Med. 2000;162(4):1277–84.CrossRefPubMed PASTEUR MC, HELLIWELL SM, HOUGHTON SJ, WEBB SC, FOWERAKER JE, COULDEN RA, et al. An investigation into causative factors in patients with Bronchiectasis. Am J Respir Crit Care Med. 2000;162(4):1277–84.CrossRefPubMed
24.
Zurück zum Zitat Davies G, Wells AU, Doffman S, Watanabe S, Wilson R. The effect of < em > Pseudomonas aeruginosa on pulmonary function in patients with bronchiectasis. Eur Respir J. 2006;28(5):974–9.CrossRefPubMed Davies G, Wells AU, Doffman S, Watanabe S, Wilson R. The effect of < em > Pseudomonas aeruginosa on pulmonary function in patients with bronchiectasis. Eur Respir J. 2006;28(5):974–9.CrossRefPubMed
25.
Zurück zum Zitat Martínez-García MA, Soler-Cataluña JJ, Perpiñá-Tordera M, Román-Sánchez P, Soriano J. Factors associated with lung function decline in adult patients with stable non-cystic fibrosis bronchiectasis. Chest. 2007;132(5):1565–72.CrossRefPubMed Martínez-García MA, Soler-Cataluña JJ, Perpiñá-Tordera M, Román-Sánchez P, Soriano J. Factors associated with lung function decline in adult patients with stable non-cystic fibrosis bronchiectasis. Chest. 2007;132(5):1565–72.CrossRefPubMed
26.
Zurück zum Zitat King PT, Holdsworth SR, Freezer NJ, Villanueva E, Holmes PW. Microbiologic follow-up study in adult bronchiectasis. Respir Med. 2007;101(8):1633–8.CrossRefPubMed King PT, Holdsworth SR, Freezer NJ, Villanueva E, Holmes PW. Microbiologic follow-up study in adult bronchiectasis. Respir Med. 2007;101(8):1633–8.CrossRefPubMed
27.
Zurück zum Zitat Ho PL, Chan KN, Ip MS, Lam WK, Ho CS, Yuen KY, et al. The effect of Pseudomonas aeruginosa infection on clinical parameters in steady-state bronchiectasis. Chest. 1998;114(6):1594–8.CrossRefPubMed Ho PL, Chan KN, Ip MS, Lam WK, Ho CS, Yuen KY, et al. The effect of Pseudomonas aeruginosa infection on clinical parameters in steady-state bronchiectasis. Chest. 1998;114(6):1594–8.CrossRefPubMed
28.
Zurück zum Zitat Lee JH, Kim YK, Kwag HJ, Chang JH. Relationships between high-resolution computed tomography, lung function and bacteriology in stable bronchiectasis. J Korean Med Sci. 2004;19(1):62–8.CrossRefPubMedPubMedCentral Lee JH, Kim YK, Kwag HJ, Chang JH. Relationships between high-resolution computed tomography, lung function and bacteriology in stable bronchiectasis. J Korean Med Sci. 2004;19(1):62–8.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Tsang KW, Rutman A, Tanaka E, Lund V, Dewar A, Cole PJ, et al. Interaction of Pseudomonas aeruginosa with human respiratory mucosa in vitro. Eur Respir J. 1994;7(10):1746–53.CrossRefPubMed Tsang KW, Rutman A, Tanaka E, Lund V, Dewar A, Cole PJ, et al. Interaction of Pseudomonas aeruginosa with human respiratory mucosa in vitro. Eur Respir J. 1994;7(10):1746–53.CrossRefPubMed
30.
Zurück zum Zitat Chalmers JD, Aliberti S, Blasi F. Management of bronchiectasis in adults. Eur Respir J. 2015;45(5):1446–62.CrossRefPubMed Chalmers JD, Aliberti S, Blasi F. Management of bronchiectasis in adults. Eur Respir J. 2015;45(5):1446–62.CrossRefPubMed
31.
Zurück zum Zitat Jen R, Rennard SI, Sin DD. Effects of inhaled corticosteroids on airway inflammation in chronic obstructive pulmonary disease: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis. 2012;7:587–95.PubMedPubMedCentral Jen R, Rennard SI, Sin DD. Effects of inhaled corticosteroids on airway inflammation in chronic obstructive pulmonary disease: a systematic review and meta-analysis. Int J Chron Obstruct Pulmon Dis. 2012;7:587–95.PubMedPubMedCentral
32.
Zurück zum Zitat Tsang KW, Ho PL, Lam WK, Ip MS, Chan KN, Ho CS, et al. Inhaled fluticasone reduces sputum inflammatory indices in severe bronchiectasis. Am J Respir Crit Care Med. 1998;158(3):723–7.CrossRefPubMed Tsang KW, Ho PL, Lam WK, Ip MS, Chan KN, Ho CS, et al. Inhaled fluticasone reduces sputum inflammatory indices in severe bronchiectasis. Am J Respir Crit Care Med. 1998;158(3):723–7.CrossRefPubMed
33.
Zurück zum Zitat Tsang KW, Chan K, Ho P, Zheng L, Ooi GC, Ho JC, et al. Sputum elastase in steady-state bronchiectasis. Chest. 2000;117(2):420–6.CrossRefPubMed Tsang KW, Chan K, Ho P, Zheng L, Ooi GC, Ho JC, et al. Sputum elastase in steady-state bronchiectasis. Chest. 2000;117(2):420–6.CrossRefPubMed
34.
Zurück zum Zitat Martínez-García MA, Perpiñá-Tordera M, Román-Sánchez P, Soler-Cataluña JJ. Inhaled steroids improve quality of life in patients with steady-state bronchiectasis. Respir Med. 2006;100(9):1623–32.CrossRefPubMed Martínez-García MA, Perpiñá-Tordera M, Román-Sánchez P, Soler-Cataluña JJ. Inhaled steroids improve quality of life in patients with steady-state bronchiectasis. Respir Med. 2006;100(9):1623–32.CrossRefPubMed
35.
Zurück zum Zitat Usmani OS, Ito K, Maneechotesuwan K, Ito M, Johnson M, Barnes PJ, et al. Glucocorticoid receptor nuclear translocation in airway cells after inhaled combination therapy. Am J Respir Crit Care Med. 2005;172(6):704–12.CrossRefPubMed Usmani OS, Ito K, Maneechotesuwan K, Ito M, Johnson M, Barnes PJ, et al. Glucocorticoid receptor nuclear translocation in airway cells after inhaled combination therapy. Am J Respir Crit Care Med. 2005;172(6):704–12.CrossRefPubMed
36.
Zurück zum Zitat Barnes PJ. Scientific rationale for inhaled combination therapy with long-acting beta2-agonists and corticosteroids. Eur Respir J. 2002;19(1):182–91.CrossRefPubMed Barnes PJ. Scientific rationale for inhaled combination therapy with long-acting beta2-agonists and corticosteroids. Eur Respir J. 2002;19(1):182–91.CrossRefPubMed
37.
Zurück zum Zitat Collins S, Caron MG, Lefkowitz RJ. Beta-adrenergic receptors in hamster smooth muscle cells are transcriptionally regulated by glucocorticoids. J Biol Chem. 1988;263(19):9067–70.CrossRefPubMed Collins S, Caron MG, Lefkowitz RJ. Beta-adrenergic receptors in hamster smooth muscle cells are transcriptionally regulated by glucocorticoids. J Biol Chem. 1988;263(19):9067–70.CrossRefPubMed
38.
Zurück zum Zitat Mak JC, Nishikawa M, Shirasaki H, Miyayasu K, Barnes PJ. Protective effects of a glucocorticoid on downregulation of pulmonary beta 2-adrenergic receptors in vivo. J Clin Investig. 1995;96(1):99–106.CrossRefPubMedPubMedCentral Mak JC, Nishikawa M, Shirasaki H, Miyayasu K, Barnes PJ. Protective effects of a glucocorticoid on downregulation of pulmonary beta 2-adrenergic receptors in vivo. J Clin Investig. 1995;96(1):99–106.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Harries TH, Rowland V, Corrigan CJ, Marshall IJ, McDonnell L, Prasad V, et al. Blood eosinophil count, a marker of inhaled corticosteroid effectiveness in preventing COPD exacerbations in post-hoc RCT and observational studies: systematic review and meta-analysis. Respir Res. 2020;21(1):3.CrossRefPubMedPubMedCentral Harries TH, Rowland V, Corrigan CJ, Marshall IJ, McDonnell L, Prasad V, et al. Blood eosinophil count, a marker of inhaled corticosteroid effectiveness in preventing COPD exacerbations in post-hoc RCT and observational studies: systematic review and meta-analysis. Respir Res. 2020;21(1):3.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Rabe KF, Martinez FJ, Ferguson GT, Wang C, Singh D, Wedzicha JA, et al. Triple inhaled therapy at two glucocorticoid doses in moderate-to-very-severe COPD. N Engl J Med. 2020;383(1):35–48.CrossRefPubMed Rabe KF, Martinez FJ, Ferguson GT, Wang C, Singh D, Wedzicha JA, et al. Triple inhaled therapy at two glucocorticoid doses in moderate-to-very-severe COPD. N Engl J Med. 2020;383(1):35–48.CrossRefPubMed
41.
Zurück zum Zitat Guan WJ, Gao YH, Xu G, Li HM, Yuan JJ, Zheng JP, et al. Bronchodilator response in adults with bronchiectasis: correlation with clinical parameters and prognostic implications. J Thorac Disease. 2016;8(1):14–23. Guan WJ, Gao YH, Xu G, Li HM, Yuan JJ, Zheng JP, et al. Bronchodilator response in adults with bronchiectasis: correlation with clinical parameters and prognostic implications. J Thorac Disease. 2016;8(1):14–23.
42.
Zurück zum Zitat Jeong HJ, Lee H, Carriere KC, Kim JH, Han JH, Shin B, et al. Effects of long-term bronchodilators in bronchiectasis patients with airflow limitation based on bronchodilator response at baseline. Int J Chron Obstruct Pulmon Dis. 2016;11:2757–64.CrossRefPubMedPubMedCentral Jeong HJ, Lee H, Carriere KC, Kim JH, Han JH, Shin B, et al. Effects of long-term bronchodilators in bronchiectasis patients with airflow limitation based on bronchodilator response at baseline. Int J Chron Obstruct Pulmon Dis. 2016;11:2757–64.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Jayaram L, Vandal AC, Chang C, Lewis C, Tong C, Tuffery C et al. Tiotropium treatment for bronchiectasis: a randomised, placebo-controlled, crossover trial. Eur Respir J. 2021. Jayaram L, Vandal AC, Chang C, Lewis C, Tong C, Tuffery C et al. Tiotropium treatment for bronchiectasis: a randomised, placebo-controlled, crossover trial. Eur Respir J. 2021.
Metadaten
Titel
Clinical outcomes of long-term inhaled combination therapies in patients with bronchiectasis and airflow obstruction
verfasst von
Hyo Jin Lee
Jung-Kyu Lee
Tae Yeon Park
Eun Young Heo
Deog Kyeom Kim
Hyun Woo Lee
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2024
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-024-02867-4

Weitere Artikel der Ausgabe 1/2024

BMC Pulmonary Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Eingreifen von Umstehenden rettet vor Erstickungstod!

15.05.2024 Fremdkörperaspiration Nachrichten

Wer sich an einem Essensrest verschluckt und um Luft ringt, benötigt vor allem rasche Hilfe. Dass Umstehende nur in jedem zweiten Erstickungsnotfall bereit waren, diese zu leisten, ist das ernüchternde Ergebnis einer Beobachtungsstudie aus Japan. Doch es gibt auch eine gute Nachricht.

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Update Innere Medizin

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