Skip to main content
Erschienen in: Respiratory Research 1/2020

Open Access 01.12.2020 | Research

Effect of high-flow nasal therapy during early pulmonary rehabilitation in patients with severe AECOPD: a randomized controlled study

verfasst von: Lan-Fang Tung, Sheng-Yeh Shen, Hui-Hsuan Shih, Yen-Ting Chen, Chia-te Yen, Shu-Chuan Ho

Erschienen in: Respiratory Research | Ausgabe 1/2020

Abstract

Background

Chronic obstructive pulmonary disease (COPD) is airway inflammation characterized and low daily physical activity. Most pulmonary rehabilitation (PR) programs are often provided to stable patients, but fewer training programs are specific for hospitalized patients with acute exacerbation (AE). Patients with AECOPD experience increased dyspnea sensations and systemic inflammation during exercise training. High-flow nasal therapy (HFNT) reduces the minute volume, lowers the respiratory rate, and decreases the work of breathing. However, it is not clear whether HFNT is efficient during exercise training. In this study, we investigated the effects of HFNT during exercise training in an early PR program among hospitalized patients with severe AECOPD.

Methods

We enrolled COPD patients hospitalized due to AE. They were randomized into two groups according to their status into HFNT PR and non-HFNT PR groups. This study collected basic data, and also assessed a pulmonary function test, 6-min walking test, blood inflammatory biomarkers, and arterial gas analysis at the baseline, and at 4 and 12 weeks of the intervention. Data were analyzed using SPSS statistical software.

Result

We recruited 44 AECOPD patients who completed the 12-week PR program. The HFNT PR program produced significant improvements in exercise tolerance as assessed by the 6-min walking distance (6MWD), reduced dyspnea sensations in the modified Medical Research Council (mMRC), and decreased systemic inflammation as evidenced by the a lower C-reactive protein (CRP) level. A reduction in the length of hospitalization was achieved with PR in the 1-year follow-up in the two groups. The HFNT PR group showed better trends of reduced air trapping in the delta inspiration capacity (IC) and an increased quality of life according to the COPD assessment test (CAT) than did the non-HFNT PR group.

Conclusions

HFNT during exercise training in early PR increases exercise tolerance and reduces systemic inflammation in hospitalized patients with severe AECOPD.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
6MWD
6-min walking distance
ABG
Arterial blood gas
AE
Acute exacerbation
ATS
American Thoracic Society
BMI
Body-mass index
BODE index
Body-mass index, degree of airway obstruction and dyspnea, and exercise capacity
BP
Blood pressure
CAT
COPD assessment test
COPD
Chronic obstructive pulmonary disease
CRP
C-reactive protein
CVD
Cardiovascular disease
FEV1
Forced expiratory volume in the first second
FVC
Forced vital capacity
GERD
Gastroesophageal reflux disease
HFNT
High-flow nasal therapy
HR
Heart rate
IC
Inspiration capacity
IL-6
Interleukin-6
IL-8
Interleukin-8
MCID
Minimal clinically important difference
mMRC
Modified Medical Research Council
NIMV
Non-invasive mechanical ventilation
PaCO2
Partial pressure of carbon dioxide
PaO2
Partial pressure of oxygen
PR
Pulmonary rehabilitation
QOL
Quality of life
RR
Respiration rate
SD
Standard deviation
WBC
White blood cells

Introduction

Chronic obstruction pulmonary disease (COPD) patients have lower daily physical activity [1], and are also characterized by low-grade systemic inflammation caused by circulating inflammatory mediators. A past study found that intracellular oxidative stress was increased in patients with severe COPD [2]. Acute exacerbation (AE) of COPD (AECOPD) is defined as episodes of acute worsening of respiratory symptoms (such as dyspnea, coughing, and sputum production) that require additional therapy [3, 4]. Treatments for AECOPD aim to minimize the negative impacts of the current exacerbation and prevent subsequent events, such as relapse or readmission to the hospital [5]. AECOPD requiring hospitalization is associated with poor outcomes, including accelerated declines in muscle strength [6] and lung function [7], a reduced health status and quality of life (QOL) [8], accelerated disease progression [9], a significant risk of recurrent exacerbations, and an increased risk of mortality [10, 11]. It was reported in the United Kingdom that 43% of patients hospitalized with AECOPD were readmitted and 12% died within 90 days. They account for more than 70% of all COPD-related costs [12], and AECOPD is responsible for patients’ clinical deterioration. In this way, treatment goals for patients with AECOPD are to minimize the negative impacts of these events and prevent their recurrence [13]. The management of severe AECOPD is divided into pharmacological (inhaled bronchodilators, steroids, and antibiotics) and non-pharmacological treatments (oxygen therapy, high-flow nasal therapy (HFNT), non-invasive mechanical ventilation (NIMV), and pulmonary rehabilitation (PR)) [5].
PR is a comprehensive non-pharmacological treatment which has the best chance of improving COPD symptoms [14]. According to an American Thoracic Society (ATS)/European Respiratory Society statement, PR is a cornerstone intervention for managing patients with stable COPD [13]. PR is a comprehensive intervention that includes exercise training, education, and behavioral changes [15]. PR benefits COPD patients at different functional stages [16] and with different phenotypes [17]. PR programs are most often provided to stable patients or after discharge [18], and fewer training programs specific to AE in unstable periods during hospitalization have been developed. Most studies that assessed the efficiency of PR during AECOPD have shown controversial results in hospitalized patients [12], who present more-severe exacerbation and/or more-severe underlying disease than stable COPD patients in an outpatient setting. The use of systemic steroids [19] and NIMV [20] for in-hospital AECOPD is supported by strong evidence of their efficacy. Some AECOPD patients are unable to reach the required training intensity to improve clinical or physiological effects. HFNT can deliver up to 60 L/min of reheated, humidified air via a nasal cannula. It can increase alveolar ventilation, improve arterial blood gas (ABG) data, and reduce the effort of breathing [2123]. HFNT in stable hypercapnia patients leads to a flow-dependent reduction in the hypercapnia level [21]. But, HFNT needs to be assessed in further prospective studies, especially when combined with exercise training and PR programs.
Therefore, this study aimed to investigate the effects of the combination of HFNT with exercise training in an early PR program in patients with AECOPD and assess its effects on hospitalization times, AECOPD patient symptoms, exercise tolerance, QOL, and functionality.

Materials and methods

Study subjects

We enrolled 54 patients with COPD who were hospitalized due to AE in Mackay Memorial Hospital (Taipei City, Taiwan) between April 2017 and April 2019; ten of them did not finish the course (Fig. 1). Patients who had received a diagnosis of COPD exhibited a post-bronchodilator forced expiratory volume in the first second (FEV1)/forced vital capacity (FVC) ratio of ≤70%. All COPD patients were in category C or D according to the combined COPD assessment using new GOLD guidelines [24]. All participants were aged 40~90 years and were ambulatory by 48 h after admission for AECOPD at the time of inclusion. Subjects are required to do every day during hospitalization, and continued twice a week until 3 months after discharge. Study subjects were then followed for 12 months. Subjects with known unstable vital signs (including a body temperature of > 38.5 °C, a respiration rate (RR) of > 40 beats/min or < 10 beats/min, and heart rate (HR) of > 150 beats/min or < 40 beats/min, and a mean blood pressure (BP) of < 70 mmHg), a malignant tumor, angina, myocardial infarction, severe hypoxemia, an unstable psychological status, hemoptysis, and pneumothorax were excluded. There was no significant adverse event during the rehabilitation program, except one participate who withdrew the program because of progressive dyspnea during lower limb exercise. All study participants provided written informed consent (17MMHIS012).

Study design

This was a prospective, cross-section, randomized controlled study in which subjects were randomized to an HFNT PR group and a non-HFNT PR group. The HFNT PR group received HFNT during exercise training in addition to the usual care and PR program. The non-HFNT PR group only received usual care and a PR program. All patients attended a 12-week, twice-a-week, in-hospital, multidisciplinary PR program, consisting of four components: (a) health education; (b) breathing exercises; (c) bronchial hygiene and lung expansion therapy; and (d) exercise training.

Health education

All patients received information from a respiratory therapist about disease awareness, the proper use of medications, pathophysiology, muscle relaxation and energy conservation methods, bronchial hygiene methods, how to cope with acute dyspnea, and nutritional guidance.

Breathing exercises

All patients were taught to use a pursed-lip/diaphragmatic breathing exercise during limb muscle training, pacing waking, and bronchial hygiene.

Bronchial hygiene

Postural drainage, percussion, vibration, and effective coughing techniques to improve airway clearance were carried out regularly daily at home and weekly at hospital visits in case of sputum impaction. At the weekly follow-up clinical visit, if needed, patients were provided with lung expansion therapy with intermittent positive pressure breathing or a negative-pressure ventilator.

Exercise training

Different types of physical exercises were used, for example, limb strength training, and ergometer bicycle training. These were constant-work-rate exercises, including warm-up, rapid-pace walking, and cool-down. At the twice-weekly follow-up clinic visits, patients underwent upper- and lower-limb bicycle ergometer training for 20 min, and perceived symptoms of breathlessness and muscle fatigue.

HFNT

The AIRVO 2 system (Fisher & Paykel, Auckland, New Zealand) was used to provide HFNT. This system generates humidified air. The flow was set to 50 L/min and provide an extended range of oxygen concentrations when needed during the exercise. The patient was instructed to breathe through the nose.

Data collection

Each participant was interviewed by a well-trained respiratory therapist to collect demographic, lifestyle, and disease-related data (smoking habits, number of exacerbations in the past year, medications used in the stable and exacerbation periods of the disease, and comorbidities) which were collected within 48 h of AECOPD onset, after the 4-week PR follow-up clinical visit, and at the end of the 12-week PR training period. After the end of the 12-week study, observational AE data were collected at a 1-year follow-up assessment. The body-mass index (BMI) was calculated as the weight divided by the height squared (kg/m2). The modified Medical Research Council (mMRC) scale, graded from 0 to 4, is a simple, valid and widely used instrument to characterize the impact of dyspnea on daily activities of patients with COPD [25, 26]. Variations of 0.6 units [27] were indicated to be a the minimal clinically important difference (MCID) for patients with AECOPD after pharmacological treatment. The impact of the disease was measured with the previously validated Chinese version of the COPD Assessment Test (CAT), and an MCID of 2 points for patients with AECOPD receiving pharmacological treatment was previously established [28].
Pulmonary function parameters were assessed using a Vitalograph Spirotac V™ (Vitalograph, England) after a 10-min rest; the post-bronchodilator FEV1 and FVC were measured, and the FEV1/FVC ratio was calculated. The exercise capacity was assessed with a 6-min walking test (6MWT), according to ATS guidelines [29]. All subjects were instructed to walk as far as possible but were allowed to stop and rest during the test. Oxygen saturation and the pulse rate were recorded using a continuous finger-adapted pulse oximeter during the 6MWT. Additionally, at the beginning and the end of the 6MWT, the difference in exertion was assessed with the modified Borg’s scale (10-point scale), for rating perceived exertion by measuring breathlessness. All patients were familiar with the 6MWT before the study.
Blood samples were collected and analyses included arterial blood gas (ABG) (pH, partial pressure of oxygen (PaO2), PaCO2 and HCO3), white blood cells (WBCs), and C-reactive protein (CRP) within 48 h after admission, at the 4-week follow-up clinical visit, and at the end of the 12-week PR training period.

Statistical analysis

Results were statistically analyzed with SPSS for Windows 19.0 (SPSS, Chicago, IL, USA) and the program Graph-Pad Prism 5 (GraphPad Software, La Jolla, CA, USA). Descriptive analyses were performed to evaluate demographic and clinical characteristics of patients. Descriptive data are expressed as the mean ± standard deviation (SD). Continuous variables were compared using a repeated-measures test, an analysis of variance (ANOVA) was used to compare data within groups, and an unpaired t-test was used to compare two groups with respect to delta mMRC, BMI, obstruction, dyspnea, exercise capacity (BODE health index: body-mass index, degree of airway obstruction and dyspnea, and exercise capacity), and CAT. Categorical variables were compared between two groups using a Chi-squared test. Statistical significance was defined as p < 0.05.

Results

Patient characteristics

In total, 44 patients finished the PR course. Twenty-two patients (15 males and seven females, mean age 73.1 ± 6.4 years; FEV1: 36.5% ± 10.3% predicted) in the non-HFNT PR group, and 22 patients (17 males and five females, mean age 72.3 ± 7.7 years; FEV1: 36.6% ± 8.0% predicted) in the HFNT PR group were enrolled. Baseline characteristics of both groups are shown in Table 1. With the exception of the smoking status, mMRC, CAT, and BODE there were no significant differences between the two groups. Patients in both the non-HFNT PR and HFNT PR groups had poor exercise tolerance (6MWD, 128.5 ± 85.6 vs. 177.0 ± 89.2 m), more-severe dyspnea sensation (mMRC, 3.5 ± 0.5 vs. 3.1 ± 0.4), and a worse health status (CAT, 30.9 ± 2.1 vs. 28.9 ± 2.8). Other features were increased respiratory infection and inflammation (WBCs, 14.8 ± 5.2 vs. 14.7 ± 4.4 103/μl; CRP, 8.4 ± 6.8 vs. 5.1 ± 5.6 mg/dl). pH 7.36 was in the normal range, but PaCO2 was above normal on the acidosis side of normal. The number of hospital admissions for AE in the previous year exceeded two times; most patients had comorbidities, especially cardiovascular diseases. The mean AE hospital length of stay was 8.46 ± 1.61 days.
Table 1
Characteristics of the patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD) patients in the high-flow nasal therapy pulmonary rehabilitation (HFNT PR) and non-HFNT PR groups
Characteristic
Non-HFNT PR
(N = 22)
HFNT PR
(N = 22)
p value
Age, years
73.1 ± 6.4
72.3 ± 7.7
0.703
Gender, M (%)
15 (77.3)
17 (77.3)
0.728#
Smoking status
  
< 0.001#
 Non-smoker, n (%)
1 (4)
3 (13)
 
 Current smoker, n (%)
5 (22)
0 (0)
 
 Ex-smoker, n (%)
16 (72)
19 (86)
 
Body-mass index, kg/m2
19.8 ± 3.1
20.3 ± 3.4
0.614
Pulmonary function test (PFT)
 FVC, % predicted normal
80.0 ± 27.8
74.3 ± 16.2
0.412
 FEV1, % predicted normal
36.5 ± 10.3
36.6 ± 8.0
0.911
 FEV1/FVC, %
39.6 ± 12.8
40.2 ± 11.0
0.851
 Delta IC, L
−0.22 ± 0.18
−0.18 ± 0.17
0.513
6MWT
 6MWD, m
128.5 ± 85.6
177.0 ± 89.2
0.0524
 SpO2 pre/post 6MWT, %
94.4/87.3
93.8/88.9
0.484/0.241
 HR pre/post 6MWT, %
98.4/125.6
96.4/ 125.6
0.572/0.979
mMRC, score
3.5 ± 0.5
3.1 ± 0.4
0.004
CAT score
30.9 ± 2.1
28.9 ± 2.8
0.009
BODE score
8.1 ± 1.3
7.1 ± 1.1
0.010
Laboratory
 WBCs, 103/μl
14.8 ± 5.2
14.7 ± 4.4
0.951
 CRP, mg/dl
8.4 ± 6.8
5.1 ± 5.6
0.086
 pH
7.36 ± 0.08
7.36 ± 0.08
0.973
 PaCO2, mmHg
51.5 ± 24.1
52.6 ± 20.3
0.8691
 PaO2, mmHg
63.2 ± 15.3
66.1 ± 14.7
0.513
 HCO3
24.9 ± 4.2
26.3 ± 6.0
0.386
AE hospitalization in the previous PR 1 year, time
2.6 ± 0.8
2.3 ± 0.7
0.167
Comorbidity
 CVD, n (%)
15 (68)
16 (72)
0.741#
 GERD, n (%)
6 (27)
9 (40)
0.340#
 Osteoporosis, n (%)
7 (31)
5 (22)
0.498#
 Diabetes, n (%)
6 (27)
5 (22)
0.728#
 Chronic renal failure, n (%)
2 (9)
3 (13)
0.635#
 Anxiety, n (%)
4 (18)
3 (13)
0.680#
Length of hospitalization, days
8.6 ± 1.3
8.3 ± 1.9
0.459
Note: Data are presented as the mean ± SD; # Analyzed by a Chi-squared test
Abbreviations: GOLD Global Initiative for Chronic Obstructive Pulmonary Lung Disease; Group D: acute exacerbation (AE) ≥ 2; modified Medical Research Council (mMRC) ≥ 2, COPD assessment test (CAT) ≥ 10. FEV1, forced expiratory volume in the first second; FVC forced vital capacity; 6MWD, 6-min walking distance; mMRC modified Medical Research Council; BODE index body-mass index, degree of airflow obstruction and dyspnea, and exercise capacity; WBCs, white blood cells; CRP, C-reactive protein; PaCO2 partial pressure of carbon dioxide; PaO2 partial pressure of oxygen; GERD gastroesophageal reflux disease; CVD cardiovascular disease

Clinical outcomes of pulmonary function, exercise tolerance, and laboratory data

In within-group comparisons of pulmonary function, FVC had significantly increased and delta IC had significantly decreased after 12 weeks in the non-HFNT PR group, and FEV1 had significantly increased after 12 weeks and delta IC had significantly decreased after 4 and 12 weeks in the HFNT PR group. By the end of the study, there was no significant difference in the pulmonary function test between the two groups (Table 2).
Table 2
Between and within groups differences on the pulmonary function test, 6-min walking test (6MWT), and laboratory data at the baseline, at 4 weeks, and after 12 weeks (N = 44)
 
Non-HFNT PR
HFNT PR
Between-group test
Baseline (n = 22)
4 weeks (n = 22)
12 weeks (n = 22)
Baseline (n = 22)
4 weeks (n = 22)
12 week (n = 22)
4 weeks
12 weeks
p value
Pulmonary function test
 FVC, L
2.29 ± 0.76
2.38 ± 0.73**a
2.40 ± 0.74**a
2.26 ± 0.68
2.38 ± 0.69
2.42 ± 0.69
0.990
0.932
 FEV1, L
0.83 ± 0.24
0.84 ± 0.23
37.5 ± 39.6
0.86 ± 0.26
0.88 ± 0.26
0.92 ± 0.26**a,b
0.550
0.324
 FEV1/FVC, %
39.6 ± 12.8
39.6 ± 12.5
39.6 ± 12.5
40.2 ± 11.0
40.3 ± 11.2
40.4 ± 11.6
0.840
0.833
 Delta IC, L
−0.22 ± 0.18
−0.18 ± 0.16
−0.16 ± 0.15*a
−0.18 ± 0.17
−0.13 ± 0.12*a
−0.09 ± 0.13**a
0.146
0.099
6MWT
 6MWD, m
128.5 ± 85.6
191.4 ± 89.3
245.5 ± 103.1**a,b
178.9 ± 81.6
260.1 ± 87.7**a
304.6 ± 84.4**a,b
0.014c
0.044c
 SpO2, % pre 6MWT
94.4 ± 3.4
95.0 ± 3.1
95.5 ± 2.6*a
93.8 ± 2.5
95.1 ± 2.4*a
95.6 ± 2.1**a
0.957
0.802
 SpO2, % post 6MWT
87.4 ± 4.2
88.9 ± 4.9*a
90.5 ± 2.8**a,b
88.9 ± 4.4
90.7 ± 3.1**a
91.8 ± 2.4**a
0.136
0.103
 HR, pre 6MWT
98.4 ± 10.7
92.1 ± 11.3**a
91.7 ± 10.8**a
96.5 ± 11.5
92.6 ± 10.5**a
91.3 ± 10.4**a
0.880
0.899
 HR, post 6MWT
125.6 ± 8.1
124.4 ± 7.8
125.1 ± 7.4
125.6 ± 14.0
122.7 ± 6.4
122.0 ± 6.6
0.426
0.141
Laboratory data
 WBCs, 103/μl
14.80 ± 5.22
8.73 ± 2.01**a
7.92 ± 1.56**a
14.71 ± 4.44
8.18 ± 2.65**a
7.06 ± 1.51**a
0.439
0.072
 CRP, mg/dl
8.41 ± 6.77
1.73 ± 1.73**a
0.30 ± 0.39**a
5.11 ± 5.60
1.07 ± 1.57**a
0.07 ± 0.12**a
0.192
0.020c
 pH
7.36 ± 0.08
7.40 ± 0.04**a
7.42 ± 0.05**a
7.36 ± 0.08
7.41 ± 0.05*a
7.42 ± 0.04**a
0.479
0.949
 PaCO2, mmHg
51.5 ± 24.1
45.1 ± 15.5*a
45.1 ± 13.5*a
52.6 ± 20.3
40.1 ± 5.5*a
46.0 ± 17.0**a
0.752
0.755
 PaO2, mmHg
63.2 ± 15.3
82.9 ± 21.0**a
81.9 ± 18.0**a
66.1 ± 14.7
82.7 ± 16.8**a
83.1 ± 17.3**a
0.976
0.825
 HCO3
24.88 ± 4.24
25.04 ± 5.82*a
27.35 ± 4.51*a,b
26.26 ± 5.98
27.15 ± 4.04
28.75 ± 4.15*a
0.169
0.290
Values are the mean ± SD; Delta inspiratory capacity (IC), post-6MWT IC - pre-6MWT IC
a Baseline vs. 4 weeks and 12 weeks; b 4 weeks vs. 12 weeks; * p < 0.05, ** p < 0.01; c delta, 12 weeks - baseline and 4 weeks - baseline
Abbreviations: FEV1 forced expiratory volume in the first second; FVC forced vital capacity; 6MWD, 6-min walking distance; SpO2 oxyhemoglobin saturation by pulse oximetry; HR heart rate; IC inspiratory capacity; WBCs white blood cells; CRP C-reactive protein; PaCO2 partial pressure of carbon dioxide; PaO2 partial pressure of oxygen; HCO3 hydrogen carbonate bicarbonate ion
For within-group comparisons, the two groups had significantly increased exercise tolerance in the 6MWD, SpO2, and HR after 4 and 12 weeks of PR. However, no significant changes were found in after exercise HR in the two groups. In between-group comparisons, the 6MWD had significantly increased after 4 and 12 weeks of the HFNT PR program (Table 2). However, no significant changes were found in the SpO2 or HR between in two groups. There was a greater increase in exercise tolerance with HFNT PR than with non-HFNT PR among AECOPD patients.
There were significantly improved clinical conditions in terms of WBCs, CRP, pH, PaCO2, and PaO2 after 4 and 12 weeks of the intervention in both the non-HFNT PR and HFNT PR groups. The non-HFNT PR group had significantly increased HCO3 levels after 4 and 12 weeks, while that of the HFNT PR group had only increased at 12 weeks. In between-group comparisons, laboratory data did not significantly differ in the two groups at 4 and 12 weeks of the two PR programs, except for CRP which had significantly decreased after 12 weeks in the HFNT PR group (Table 2). AECOPD patients who underwent HFNT PR had a greater decrease in the inflammation level compared to AECOPD patients in the non-HFNT PR group.

Dyspnea sensation

In within-group comparisons, the non-HFNT PR and HFNT PR groups had significantly decreased dyspnea sensations according to the mMRC after 4 and 12 weeks of the intervention (Fig. 2a). Delta mMRC had not significantly increased at 4 weeks (− 0.546 ± 0.510, − 0.409 ± 0.503) of the PR intervention in the between-group comparisons, and only delta mMRC had significantly deceased at 12 weeks (− 1.091 ± 0.294, − 0.818 ± 0.395) (Fig. 2b).

QOL

In within-group comparisons, the non-HFNT PR and HFNT PR groups had significantly improved health statuses according to CAT and BODE scores after 4 and 12 weeks of PR (Fig. 3a, c). The delta CAT (4 weeks: − 4.955 ± 2.104, − 5.545 ± 2.988; 12 weeks: − 8.500 ± 3.203, − 9.955 ± 2.984) and BODE (4 weeks: − 1.136 ± 0.710, − 1.045 ± 0.785; 12 weeks: − 2.045 ± 0.722, − 1.955 ± 0.844) scores exhibited no significant differences in between-group comparisons at 4 or 12 weeks of PR (Fig. 3b, d).

Hospital admissions for AE

Hospital admissions for AE were compared in the previous year and at 1 year after the PR program (2.43 ± 0.76 vs. 0.50 ± 0.74 times/year), and results showed a significant reduction in the number of admissions (Fig. 4a). In within-group comparisons, the non-HFNT PR (2.59 ± 0.80 vs. 0.60 ± 0.82 times/year) and HFNT PR groups (2.27 ± 0.70 vs. 0.41 ± 0.67 times/year) had significantly decreased AE times at 1 year after the PR intervention, and no significant change was found in the between-group comparisons (Fig. 4b).

Discussion

This study showed some significant outcomes after HFNT early PR in patients with AECOPD: improved exercise tolerance in the 6MWD after 4 and 12 weeks of the PR program, and decreased dyspnea sensation in mMRC and inflammation as evidenced by the CRP level after 12 weeks of PR. Moreover, a reduction in the length of hospitalization was achieved with PR at the 1-year follow-up; HFNT PR patients showed even better trends in QOL, reduced air trapping, and lower lung hyperinflation than non-HFNT PR patients, but the differences were not statistically significant between the two groups.
The first evidence of PR in acute respiratory patients was published by Trooster and colleagues: AECOPD patients who received training showed improvements in quadriceps force and 6MWD at discharge, and improvements were also documented at 1 month of follow-up [30]. The 6MWD is accepted as a good outcome measure after interventions such as PR [31, 32]. Our study had similar findings of HFNT PR patients exhibiting improved exercise tolerance as assessed by the 6MWD than non-HFNT PR patients after 4 and 12 weeks of PR. This opens up new possibilities for early rehabilitation treatment of patients hospitalized for AECOPD [33]. A systematic review suggested that PR after COPD exacerbation may reduce hospital admissions and mortality, and may improve the health-related QOL [34]. Our study reports decreased AE hospital admissions and an improved QOL including in the physical and functional domains from BODE within the two PR groups, although there were no significant differences in between-group comparisons.
The BODE index characterizes a multistage functional scoring system for COPD by means of a simple scale and requires no special equipment [35, 36]. Symptoms are the cornerstone for diagnosing AE of COPD. Dyspnea represents the most disabling symptom of COPD; the mMRC dyspnea scale is simple to administer and correlates with scores of health status [37]. In this study, after PR there were significantly decreased mMRC dyspnea levels at 4 and 12 weeks, and most patients improved above the MCID in the mMRC. Although HFNT PR significant ameliorated airflow obstruction in terms of FEV1 at 12 weeks, there were no significant differences in between-group comparisons, which is probably related to the small sample size and indicates the need for longer-term follow-up.
CAT impacts a patient’s QOL [38], is responsive to treatment [39], and provides relevant prognostic information [40]; therefore, its use is advocated for assessing PR during AECOPD. In our study, after PR, there were significant improvements in the QOL at 4 and 12 weeks, and most patients improved above the MCID in CAT, although there were no significant differences in between-group comparisons.
Lung hyperinflation limits the expiratory flow in patients with COPD and contributes to dyspnea and activity limitations [41]; it has become an important therapeutic target in symptomatic COPD patients [42]. In our study, respiratory therapists were taught to use the pulse-lip/diaphragm breathing technique, relaxation exercises, and combined HFNT during exercise. HFNT patients showed better trends of reduced air trapping in delta IC after exercise compared to non-HFNT PR patients.
HFNT provides warmed humidified air administered through slightly enlarged nasal prongs. HFNT reduces the minute volume, lowers the respiratory rate, and decreases the work of breathing [21, 43], and it leads to a flow-dependent reduction in PaCO2 [21]. When we analyzed data on arterial blood gases (pH, PaO2, PaCO2, and HCO3−), our results of reduced PaCO2 levels were similar within each group, but did not significantly differ between the two groups at the end of the PR program. It is well known that AECOPD may be triggered by infection with bacteria or viruses or by noninfectious environmental or internal factors. Patients with AECOPD also display heterogeneous inflammation, and inflammatory markers such as CRP [44] as a kind of quantitative indicator are widely used in judging AE and for assessing prognoses [45]. Serum CRP may provide prognostic information about morbidity and mortality in COPD patients because of relationships among CRP, interleukin (IL)-6, exercise tolerance, and the health status [46, 47]. Our study proved that an HFNT PR program reduced CRP levels better than a non-HFNT PR program, and utilizing HFNT during exercise in the COPD PR program might help avoid fatigue, decrease exercise limitations related to the excessive load placed on inspiratory muscles, and reduce systemic inflammation.
Our study had some limitations. First, there was a small simple size, and only one hospital participated in the study. Second, too few women were recruited, and recent data indicate that female COPD patients may have a higher number of hospitalizations with a more-prolonged length of stay [48]. Third, pharmacological treatment was not standardized, although there were no differences at the baseline assessment, it must be acknowledged that this might have influenced patients’ recovery. Fourth, correlations of laboratory blood samples were not assessed for COPD inflammatory cytokines and oxidative markers such as IL-8, TNF-α, 8-isoprostant, and so on.

Conclusions

High-flow nasal therapy during exercise training in early pulmonary rehabilitation is feasible for hospitalized patients with severe AECOPD who are profoundly intolerant of exercise training. Exercise training with HFNT can support an increased exercise capacity, decreased dyspnea sensations, and reductions in systemic inflammatory biomarkers in hospitalized patients with severe AECOPD. Our study also raises the possibility that HFNT during exercise enhances the training effect through decreased lung hyperinflation and increased pulmonary function.

Acknowledgments

The authors wish to thank the patients and personnel of the hospital unit for their cooperation during the course of this study.
The experimental protocol was established, according to the ethical guidelines of the Helsinki Declaration and was approved by the Human Ethics Committee of Mackay Memorial Hospital Institutional. Written informed consent was obtained from individual or guardian participants.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis 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.
Literatur
1.
Zurück zum Zitat Lee KY, Chen TT, Chiang LL, Chuang HC, Feng PH, Liu WT, Chen KY, Ho SC. Proteasome activity related with the daily physical activity of COPD patients. Int J Chron Obstruct Pulmon Dis. 2017;12:1519–25.PubMedPubMedCentralCrossRef Lee KY, Chen TT, Chiang LL, Chuang HC, Feng PH, Liu WT, Chen KY, Ho SC. Proteasome activity related with the daily physical activity of COPD patients. Int J Chron Obstruct Pulmon Dis. 2017;12:1519–25.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Lee KY, Ho SC, Chan YF, Wang CH, Huang CD, Liu WT, Lin SM, Lo YL, Chang YL, Kuo LW, Kuo HP. Reduced nuclear factor-kappaB repressing factor: a link toward systemic inflammation in COPD. Eur Respir J. 2012;40:863–73.PubMedCrossRef Lee KY, Ho SC, Chan YF, Wang CH, Huang CD, Liu WT, Lin SM, Lo YL, Chang YL, Kuo LW, Kuo HP. Reduced nuclear factor-kappaB repressing factor: a link toward systemic inflammation in COPD. Eur Respir J. 2012;40:863–73.PubMedCrossRef
3.
Zurück zum Zitat Wedzicha JAEC-C, Miravitlles M, Hurst JR, Calverley PM, Albert RK, Anzueto A, Criner GJ, Papi A, Rabe KF, Rigau D, et al. Management of COPD exacerbations: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2017;49:1600791. Wedzicha JAEC-C, Miravitlles M, Hurst JR, Calverley PM, Albert RK, Anzueto A, Criner GJ, Papi A, Rabe KF, Rigau D, et al. Management of COPD exacerbations: a European Respiratory Society/American Thoracic Society guideline. Eur Respir J. 2017;49:1600791.
4.
Zurück zum Zitat Cavailles A, Brinchault-Rabin G, Dixmier A, Goupil F, Gut-Gobert C, Marchand-Adam S, Meurice JC, Morel H, Person-Tacnet C, Leroyer C, Diot P. Comorbidities of COPD. Eur Respir Rev. 2013;22:454–75.PubMedCrossRef Cavailles A, Brinchault-Rabin G, Dixmier A, Goupil F, Gut-Gobert C, Marchand-Adam S, Meurice JC, Morel H, Person-Tacnet C, Leroyer C, Diot P. Comorbidities of COPD. Eur Respir Rev. 2013;22:454–75.PubMedCrossRef
5.
Zurück zum Zitat Crisafulli E, Barbeta E, Ielpo A, Torres A. Management of severe acute exacerbations of COPD: an updated narrative review. Multidiscip Respir Med. 2018;13:36.PubMedPubMedCentralCrossRef Crisafulli E, Barbeta E, Ielpo A, Torres A. Management of severe acute exacerbations of COPD: an updated narrative review. Multidiscip Respir Med. 2018;13:36.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Spruit MA, Gosselink R, Troosters T, Kasran A, Gayan-Ramirez G, Bogaerts P, Bouillon R, Decramer M. Muscle force during an acute exacerbation in hospitalised patients with COPD and its relationship with CXCL8 and IGF-I. Thorax. 2003;58:752–6.PubMedPubMedCentralCrossRef Spruit MA, Gosselink R, Troosters T, Kasran A, Gayan-Ramirez G, Bogaerts P, Bouillon R, Decramer M. Muscle force during an acute exacerbation in hospitalised patients with COPD and its relationship with CXCL8 and IGF-I. Thorax. 2003;58:752–6.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Cote CG, Dordelly LJ, Celli BR. Impact of COPD exacerbations on patient-centered outcomes. Chest. 2007;131:696–704.PubMedCrossRef Cote CG, Dordelly LJ, Celli BR. Impact of COPD exacerbations on patient-centered outcomes. Chest. 2007;131:696–704.PubMedCrossRef
8.
Zurück zum Zitat Schmier JK, Halpern MT, Higashi MK, Bakst A. The quality of life impact of acute exacerbations of chronic bronchitis (AECB): a literature review. Qual Life Res. 2005;14:329–47.PubMedCrossRef Schmier JK, Halpern MT, Higashi MK, Bakst A. The quality of life impact of acute exacerbations of chronic bronchitis (AECB): a literature review. Qual Life Res. 2005;14:329–47.PubMedCrossRef
9.
Zurück zum Zitat Seemungal TA, Donaldson GC, Paul EA, Bestall JC, Jeffries DJ, Wedzicha JA. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418–22.PubMedCrossRef Seemungal TA, Donaldson GC, Paul EA, Bestall JC, Jeffries DJ, Wedzicha JA. Effect of exacerbation on quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:1418–22.PubMedCrossRef
10.
Zurück zum Zitat Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847–52.PubMedPubMedCentralCrossRef Donaldson GC, Seemungal TA, Bhowmik A, Wedzicha JA. Relationship between exacerbation frequency and lung function decline in chronic obstructive pulmonary disease. Thorax. 2002;57:847–52.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Suissa S, Dell'Aniello S, Ernst P. Long-term natural history of chronic obstructive pulmonary disease: severe exacerbations and mortality. Thorax. 2012;67:957–63.PubMedCrossRef Suissa S, Dell'Aniello S, Ernst P. Long-term natural history of chronic obstructive pulmonary disease: severe exacerbations and mortality. Thorax. 2012;67:957–63.PubMedCrossRef
12.
Zurück zum Zitat Puhan MA, Gimeno-Santos E, Cates CJ, Troosters T. Pulmonary rehabilitation following exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2016;12:CD005305.PubMed Puhan MA, Gimeno-Santos E, Cates CJ, Troosters T. Pulmonary rehabilitation following exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2016;12:CD005305.PubMed
13.
Zurück zum Zitat Machado A, Oliveira A, Valente C, Burtin C, Marques A. Effects of a community-based pulmonary rehabilitation programme during acute exacerbations of chronic obstructive pulmonary disease - a quasi-experimental pilot study. Pulmonology. 2019. Machado A, Oliveira A, Valente C, Burtin C, Marques A. Effects of a community-based pulmonary rehabilitation programme during acute exacerbations of chronic obstructive pulmonary disease - a quasi-experimental pilot study. Pulmonology. 2019.
14.
Zurück zum Zitat Spruit MA, Singh SJ, Garvey C, ZuWallack R, Nici L, Rochester C, Hill K, Holland AE, Lareau SC, Man WD, et al. An official American Thoracic Society/European Respiratory Society statement: key concepts and advances in pulmonary rehabilitation. Am J Respir Crit Care Med. 2013;188:e13–64.PubMedCrossRef Spruit MA, Singh SJ, Garvey C, ZuWallack R, Nici L, Rochester C, Hill K, Holland AE, Lareau SC, Man WD, et al. An official American Thoracic Society/European Respiratory Society statement: key concepts and advances in pulmonary rehabilitation. Am J Respir Crit Care Med. 2013;188:e13–64.PubMedCrossRef
15.
Zurück zum Zitat Singh SJ, ZuWallack RL, Garvey C, Spruit MA. American Thoracic Society/European Respiratory Society Task Force on Pulmonary R: Learn from the past and create the future: the 2013 ATS/ERS statement on pulmonary rehabilitation. Eur Respir J. 2013(42):1169–74. Singh SJ, ZuWallack RL, Garvey C, Spruit MA. American Thoracic Society/European Respiratory Society Task Force on Pulmonary R: Learn from the past and create the future: the 2013 ATS/ERS statement on pulmonary rehabilitation. Eur Respir J. 2013(42):1169–74.
16.
Zurück zum Zitat Vogiatzis I, Terzis G, Stratakos G, Cherouveim E, Athanasopoulos D, Spetsioti S, Nasis I, Manta P, Roussos C, Zakynthinos S. Effect of pulmonary rehabilitation on peripheral muscle fiber remodeling in patients with COPD in GOLD stages II to IV. Chest. 2011;140:744–52.PubMedCrossRef Vogiatzis I, Terzis G, Stratakos G, Cherouveim E, Athanasopoulos D, Spetsioti S, Nasis I, Manta P, Roussos C, Zakynthinos S. Effect of pulmonary rehabilitation on peripheral muscle fiber remodeling in patients with COPD in GOLD stages II to IV. Chest. 2011;140:744–52.PubMedCrossRef
17.
Zurück zum Zitat Crisafulli E, Venturelli E, Biscione G, Vagheggini G, Iattoni A, Lucic S, Ambrosino N, Pasqua F, Cesario A, Clini EM. Exercise performance after standard rehabilitation in COPD patients with lung hyperinflation. Intern Emerg Med. 2014;9:23–31.PubMedCrossRef Crisafulli E, Venturelli E, Biscione G, Vagheggini G, Iattoni A, Lucic S, Ambrosino N, Pasqua F, Cesario A, Clini EM. Exercise performance after standard rehabilitation in COPD patients with lung hyperinflation. Intern Emerg Med. 2014;9:23–31.PubMedCrossRef
18.
Zurück zum Zitat Seymour JM, Moore L, Jolley CJ, Ward K, Creasey J, Steier JS, Yung B, Man WD, Hart N, Polkey MI, Moxham J. Outpatient pulmonary rehabilitation following acute exacerbations of COPD. Thorax. 2010;65:423–8.PubMedCrossRef Seymour JM, Moore L, Jolley CJ, Ward K, Creasey J, Steier JS, Yung B, Man WD, Hart N, Polkey MI, Moxham J. Outpatient pulmonary rehabilitation following acute exacerbations of COPD. Thorax. 2010;65:423–8.PubMedCrossRef
19.
Zurück zum Zitat Davies L, Nisar M, Pearson MG, Costello RW, Earis JE, Calverley PM. Oral corticosteroid trials in the management of stable chronic obstructive pulmonary disease. QJM. 1999;92:395–400.PubMedCrossRef Davies L, Nisar M, Pearson MG, Costello RW, Earis JE, Calverley PM. Oral corticosteroid trials in the management of stable chronic obstructive pulmonary disease. QJM. 1999;92:395–400.PubMedCrossRef
20.
Zurück zum Zitat Brochard L, Mancebo J, Wysocki M, Lofaso F, Conti G, Rauss A, Simonneau G, Benito S, Gasparetto A, Lemaire F, et al. Noninvasive ventilation for acute exacerbations of chronic obstructive pulmonary disease. N Engl J Med. 1995;333:817–22.PubMedCrossRef Brochard L, Mancebo J, Wysocki M, Lofaso F, Conti G, Rauss A, Simonneau G, Benito S, Gasparetto A, Lemaire F, et al. Noninvasive ventilation for acute exacerbations of chronic obstructive pulmonary disease. N Engl J Med. 1995;333:817–22.PubMedCrossRef
21.
22.
Zurück zum Zitat Fraser JF, Spooner AJ, Dunster KR, Anstey CM, Corley A. Nasal high flow oxygen therapy in patients with COPD reduces respiratory rate and tissue carbon dioxide while increasing tidal and end-expiratory lung volumes: a randomised crossover trial. Thorax. 2016;71:759–61.PubMedCrossRef Fraser JF, Spooner AJ, Dunster KR, Anstey CM, Corley A. Nasal high flow oxygen therapy in patients with COPD reduces respiratory rate and tissue carbon dioxide while increasing tidal and end-expiratory lung volumes: a randomised crossover trial. Thorax. 2016;71:759–61.PubMedCrossRef
23.
Zurück zum Zitat Biselli PJ, Kirkness JP, Grote L, Fricke K, Schwartz AR, Smith P, Schneider H. Nasal high-flow therapy reduces work of breathing compared with oxygen during sleep in COPD and smoking controls: a prospective observational study. J Appl Physiol. 2017;122(1985):82–8.PubMedCrossRef Biselli PJ, Kirkness JP, Grote L, Fricke K, Schwartz AR, Smith P, Schneider H. Nasal high-flow therapy reduces work of breathing compared with oxygen during sleep in COPD and smoking controls: a prospective observational study. J Appl Physiol. 2017;122(1985):82–8.PubMedCrossRef
24.
Zurück zum Zitat Mirza S, Clay RD, Koslow MA, Scanlon PD. COPD Guidelines: A Review of the 2018 GOLD Report. Mayo Clin Proc. 2018(93):1488–502. Mirza S, Clay RD, Koslow MA, Scanlon PD. COPD Guidelines: A Review of the 2018 GOLD Report. Mayo Clin Proc. 2018(93):1488–502.
25.
Zurück zum Zitat Han MK, Muellerova H, Curran-Everett D, Dransfield MT, Washko GR, Regan EA, Bowler RP, Beaty TH, Hokanson JE, Lynch DA, et al: GOLD 2011 Disease severity classification in COPDGene: a prospective cohort study. Lancet Respir Med 2013, 1:43–50. Han MK, Muellerova H, Curran-Everett D, Dransfield MT, Washko GR, Regan EA, Bowler RP, Beaty TH, Hokanson JE, Lynch DA, et al: GOLD 2011 Disease severity classification in COPDGene: a prospective cohort study. Lancet Respir Med 2013, 1:43–50.
26.
Zurück zum Zitat Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Criner GJ, Frith P, Halpin DMG, Han M, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease: the GOLD science committee report. Eur Respir J. 2019;2019:53. Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Criner GJ, Frith P, Halpin DMG, Han M, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease: the GOLD science committee report. Eur Respir J. 2019;2019:53.
27.
Zurück zum Zitat Jones PW, Harding G, Berry P, Wiklund I, Chen WH, Kline Leidy N. Development and first validation of the COPD assessment test. Eur Respir J. 2009;34:648–54.PubMedCrossRef Jones PW, Harding G, Berry P, Wiklund I, Chen WH, Kline Leidy N. Development and first validation of the COPD assessment test. Eur Respir J. 2009;34:648–54.PubMedCrossRef
28.
Zurück zum Zitat Kon SS, Canavan JL, Jones SE, Nolan CM, Clark AL, Dickson MJ, Haselden BM, Polkey MI, Man WD. Minimum clinically important difference for the COPD assessment test: a prospective analysis. Lancet Respir Med. 2014;2:195–203.PubMedCrossRef Kon SS, Canavan JL, Jones SE, Nolan CM, Clark AL, Dickson MJ, Haselden BM, Polkey MI, Man WD. Minimum clinically important difference for the COPD assessment test: a prospective analysis. Lancet Respir Med. 2014;2:195–203.PubMedCrossRef
29.
Zurück zum Zitat Laboratories ATSCoPSfCPF. ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med. 2002;166:111–7.CrossRef Laboratories ATSCoPSfCPF. ATS statement: guidelines for the six-minute walk test. Am J Respir Crit Care Med. 2002;166:111–7.CrossRef
30.
Zurück zum Zitat Troosters T, Probst VS, Crul T, Pitta F, Gayan-Ramirez G, Decramer M, Gosselink R. Resistance training prevents deterioration in quadriceps muscle function during acute exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181:1072–7.PubMedCrossRef Troosters T, Probst VS, Crul T, Pitta F, Gayan-Ramirez G, Decramer M, Gosselink R. Resistance training prevents deterioration in quadriceps muscle function during acute exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181:1072–7.PubMedCrossRef
31.
Zurück zum Zitat Pulmonary rehabilitation-1999. American Thoracic Society. Am J Respir Crit Care Med. 1999;159:1666–82. Pulmonary rehabilitation-1999. American Thoracic Society. Am J Respir Crit Care Med. 1999;159:1666–82.
32.
Zurück zum Zitat Enright PL. The six-minute walk test. Respir Care. 2003;48:783–5.PubMed Enright PL. The six-minute walk test. Respir Care. 2003;48:783–5.PubMed
33.
Zurück zum Zitat Clini E, Roversi P, Crisafulli E. Early rehabilitation: much better than nothing. Am J Respir Crit Care Med. 2010;181:1016–7.PubMedCrossRef Clini E, Roversi P, Crisafulli E. Early rehabilitation: much better than nothing. Am J Respir Crit Care Med. 2010;181:1016–7.PubMedCrossRef
34.
Zurück zum Zitat Puhan M, Scharplatz M, Troosters T, Walters EH, Steurer J. Pulmonary rehabilitation following exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2009:CD005305. Puhan M, Scharplatz M, Troosters T, Walters EH, Steurer J. Pulmonary rehabilitation following exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2009:CD005305.
35.
Zurück zum Zitat Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M, Mendez RA, Pinto Plata V, Cabral HJ. the body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:1005–12.CrossRefPubMed Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M, Mendez RA, Pinto Plata V, Cabral HJ. the body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:1005–12.CrossRefPubMed
36.
Zurück zum Zitat Funk GC, Kirchheiner K, Burghuber OC, Hartl S. BODE index versus GOLD classification for explaining anxious and depressive symptoms in patients with COPD - a cross-sectional study. Respir Res. 2009;10:1.PubMedPubMedCentralCrossRef Funk GC, Kirchheiner K, Burghuber OC, Hartl S. BODE index versus GOLD classification for explaining anxious and depressive symptoms in patients with COPD - a cross-sectional study. Respir Res. 2009;10:1.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Hajiro T, Nishimura K, Tsukino M, Ikeda A, Koyama H, Izumi T. Comparison of discriminative properties among disease-specific questionnaires for measuring health-related quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:785–90.PubMedCrossRef Hajiro T, Nishimura K, Tsukino M, Ikeda A, Koyama H, Izumi T. Comparison of discriminative properties among disease-specific questionnaires for measuring health-related quality of life in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157:785–90.PubMedCrossRef
38.
Zurück zum Zitat Haughney J, Partridge MR, Vogelmeier C, Larsson T, Kessler R, Stahl E, Brice R, Lofdahl CG. Exacerbations of COPD: quantifying the patient's perspective using discrete choice modelling. Eur Respir J. 2005;26:623–9.PubMedCrossRef Haughney J, Partridge MR, Vogelmeier C, Larsson T, Kessler R, Stahl E, Brice R, Lofdahl CG. Exacerbations of COPD: quantifying the patient's perspective using discrete choice modelling. Eur Respir J. 2005;26:623–9.PubMedCrossRef
39.
Zurück zum Zitat Mackay AJ, Donaldson GC, Patel AR, Jones PW, Hurst JR, Wedzicha JA. Usefulness of the chronic obstructive pulmonary disease assessment test to evaluate severity of COPD exacerbations. Am J Respir Crit Care Med. 2012;185:1218–24.PubMedCrossRef Mackay AJ, Donaldson GC, Patel AR, Jones PW, Hurst JR, Wedzicha JA. Usefulness of the chronic obstructive pulmonary disease assessment test to evaluate severity of COPD exacerbations. Am J Respir Crit Care Med. 2012;185:1218–24.PubMedCrossRef
40.
Zurück zum Zitat Garcia-Sidro P, Naval E, Martinez Rivera C, Bonnin-Vilaplana M, Garcia-Rivero JL, Herrejon A, Malo de Molina R, Marcos PJ, Mayoralas-Alises S, Ros JA, et al. the CAT (COPD assessment test) questionnaire as a predictor of the evolution of severe COPD exacerbations. Respir Med. 2015;109:1546–52.PubMedCrossRef Garcia-Sidro P, Naval E, Martinez Rivera C, Bonnin-Vilaplana M, Garcia-Rivero JL, Herrejon A, Malo de Molina R, Marcos PJ, Mayoralas-Alises S, Ros JA, et al. the CAT (COPD assessment test) questionnaire as a predictor of the evolution of severe COPD exacerbations. Respir Med. 2015;109:1546–52.PubMedCrossRef
41.
Zurück zum Zitat Aliverti A, Ghidoli G, Dellaca RL, Pedotti A, Macklem PT. Chest wall kinematic determinants of diaphragm length by optoelectronic plethysmography and ultrasonography. J Appl Physiol. 2003;94(1985):621–30.PubMedCrossRef Aliverti A, Ghidoli G, Dellaca RL, Pedotti A, Macklem PT. Chest wall kinematic determinants of diaphragm length by optoelectronic plethysmography and ultrasonography. J Appl Physiol. 2003;94(1985):621–30.PubMedCrossRef
42.
Zurück zum Zitat O'Donnell DE, Revill SM, Webb KA. Dynamic hyperinflation and exercise intolerance in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:770–7.PubMedCrossRef O'Donnell DE, Revill SM, Webb KA. Dynamic hyperinflation and exercise intolerance in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001;164:770–7.PubMedCrossRef
43.
Zurück zum Zitat Delorme M, Bouchard PA, Simon M, Simard S, Lellouche F. Effects of high-flow nasal cannula on the work of breathing in patients recovering from acute respiratory failure. Crit Care Med. 2017;45:1981–8.PubMedCrossRef Delorme M, Bouchard PA, Simon M, Simard S, Lellouche F. Effects of high-flow nasal cannula on the work of breathing in patients recovering from acute respiratory failure. Crit Care Med. 2017;45:1981–8.PubMedCrossRef
44.
Zurück zum Zitat Karadeniz G, Polat G, Senol G, Buyuksirin M. C-reactive protein measurements as a marker of the severity of chronic obstructive pulmonary disease exacerbations. Inflammation. 2013;36:948–53.PubMedCrossRef Karadeniz G, Polat G, Senol G, Buyuksirin M. C-reactive protein measurements as a marker of the severity of chronic obstructive pulmonary disease exacerbations. Inflammation. 2013;36:948–53.PubMedCrossRef
45.
Zurück zum Zitat Zhou A, Zhou Z, Zhao Y, Chen P. The recent advances of phenotypes in acute exacerbations of COPD. Int J Chron Obstruct Pulmon Dis. 2017;12:1009–18.PubMedPubMedCentralCrossRef Zhou A, Zhou Z, Zhao Y, Chen P. The recent advances of phenotypes in acute exacerbations of COPD. Int J Chron Obstruct Pulmon Dis. 2017;12:1009–18.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Man SF, Connett JE, Anthonisen NR, Wise RA, Tashkin DP, Sin DD. C-reactive protein and mortality in mild to moderate chronic obstructive pulmonary disease. Thorax. 2006;61:849–53.PubMedPubMedCentralCrossRef Man SF, Connett JE, Anthonisen NR, Wise RA, Tashkin DP, Sin DD. C-reactive protein and mortality in mild to moderate chronic obstructive pulmonary disease. Thorax. 2006;61:849–53.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Garcia-Rio F, Miravitlles M, Soriano JB, Munoz L, Duran-Tauleria E, Sanchez G, Sobradillo V, Ancochea J, Committee E-SS. Systemic inflammation in chronic obstructive pulmonary disease: a population-based study. Respir Res. 2010;11:63.PubMedPubMedCentralCrossRef Garcia-Rio F, Miravitlles M, Soriano JB, Munoz L, Duran-Tauleria E, Sanchez G, Sobradillo V, Ancochea J, Committee E-SS. Systemic inflammation in chronic obstructive pulmonary disease: a population-based study. Respir Res. 2010;11:63.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Kilic H, Kokturk N, Sari G, Cakir M. Do females behave differently in COPD exacerbation? Int J Chron Obstruct Pulmon Dis. 2015;10:823–30.PubMedPubMedCentral Kilic H, Kokturk N, Sari G, Cakir M. Do females behave differently in COPD exacerbation? Int J Chron Obstruct Pulmon Dis. 2015;10:823–30.PubMedPubMedCentral
Metadaten
Titel
Effect of high-flow nasal therapy during early pulmonary rehabilitation in patients with severe AECOPD: a randomized controlled study
verfasst von
Lan-Fang Tung
Sheng-Yeh Shen
Hui-Hsuan Shih
Yen-Ting Chen
Chia-te Yen
Shu-Chuan Ho
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2020
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-020-1328-z

Weitere Artikel der Ausgabe 1/2020

Respiratory Research 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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