Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2019

Open Access 01.12.2019 | Research article

Acute exacerbations of fibrosing interstitial lung disease associated with connective tissue diseases: a population-based study

verfasst von: Mengshu Cao, Jian Sheng, Xiaohua Qiu, Dandan Wang, Dongmei Wang, Yang Wang, Yonglong Xiao, Hourong Cai

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2019

Abstract

Background

Acute exacerbation (AE) is the major cause of morbidity and mortality in patients with idiopathic pulmonary fibrosis (IPF). AEs also occur in other forms of fibrosing interstitial lung disease (fILD). The clinical features and prognosis of AE patients with connective tissue diseases (CTDs) associated-ILD has not been fully described.

Methods

We retrospectively reviewed 177 patients with either IPF or a characterized CTD-ILD admitted to Nanjing Drum Tower Hospital with an AE from January 2010 to December 2016.

Results

The study cohort included 107 subjects with AE-IPF and 70 cases with AE-CTD-ILD. Female gender, prior use of corticosteroid and immunosupressants, lower serum albumin, higher D-dimer level, TLC% pred, survival, and treatment with immunosupressants and caspofungin were more common in the CTD-ILD group (all p<0.05). The incidences of AE-CTD-ILD and AE-IPF were similar in our single center (p = 0.526). TLC% pred was the risk factor for AE after ILD diagnosis for 1 year in CTD patients (p = 0.018). Log-rank tests showed patients with CTD-ILD had a significantly lower mortality rate compared with IPF patients after AEs (p = 0.029). No significant difference in survival was noted among CTD subgroups (p = 0.353). The survival was negatively correlated with WBC count, LDH and CT score, (p = 0.006, p = 0.013 and p = 0.035, respectively), and positively correlated with PaO2/FiO2 ratio (p<0.001) in the CTD-ILD group. WBC count and PO2/FiO2 ratio were the independent predictors for survival in AE-CTD-ILD after adjusting for other clinical variates in Cox regression Models (p = 0.038 and p < 0.001, respectively).

Conclusions

The clinical characteristics of patients with AE-CTD-ILD differed from those with AE-IPF, while AE incidences were similar between the two groups. Subjects with AE-CTD-fILD tended to have a better prognosis, and WBC count and PO2/FiO2 ratio were the independent survival predictors for these patients.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12890-019-0960-1.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AE
Acute exacerbation
ALB
Albumin
BNP
B-type natriuretic peptide
CRP
C reactive protein
CTDs
Connective tissue diseases
DLCO
Diffusing capacity of the lung for CO2
ESR
Erythrocyte sedimentation rate
fILD
Fibrosing interstitial lung disease
FVC
Forced vital capacity
HRCT
High resolution computed tomography
IPF
Idiopathic pulmonary fibrosis
LDH
Lactate dehydrogenase
MV
Mechanical ventilation
NAC
N-acetylcysteine
PAH
Pulmonary arterial hypertension; BMI: body mass index
PaO2/FiO2
Oxygenation index
P-UIP
Possible UIP
TLC
Total lung capacity
UIP
Usual interstitial pneumonia
WBC
White blood cell

Background

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive scarring disease of the lung characterized by worsening dyspnea and lung function over time [1, 2]. Some patients will experience an acute, clinically significant respiratory deterioration characterized by widespread alveolar abnormality [3]. Acute exacerbations (AEs) are a major cause of mortality of patients with IPF. The incidence of AE-IPF ranges from 4 to 20% per year among IPF patients in reported clinical trials [48], and up to one-half of IPF patients die from AE [9]. In most cases, in-hospital mortality rate was up to 50% [7]. The etiology of AE-IPF is still unknown. Recent evidence suggests that causes of acute lung injury (ALI) and histopathologic diffuse alveolar damage (DAD) such as infection, aspiration, surgery and air pollution exposure could contribute to the development of AE-IPF [3, 10, 11]. Low forced vital capacity (FVC), diffusing capacity for carbonmonoxide (DLCO) and baseline oxygenation were known risk factors for AE [7, 10, 1214]. Lower baseline FVC and DLCO and more extensive CT abnormalities at the time of AE and worse oxygenation were predictors of mortality [7, 8, 15]. The elevated levels of Kerbs von Lungen-6 and leptin in peripheral blood could be the risk and prognostic factors for AE-IPF [16, 17].
Recently, AE has also been shown to occur in fibrosing interstitial lung diseases (fILD) other than IPF, such as connective tissue disease (CTD) associated ILD, idiopathic nonspecific interstitial lung disease and chronic hypersensitivity pneumonias [1822]. Park et al. reported AE in CTD-ILD was similar to AE-IPF and most common in older patients with rheumatoid arthritis (RA) [18]. Suda et al. suggested that AE occurred mostly with RA-ILD with a usual interstitial pneumonia (UIP) pattern [19]. Toyoda et al. showed some AE cases in patients with CTD had good response to corticosteroid [23]. To date, there are few reports of AE in CTD-ILD patients. The incidence, clinical characteristics and prognosis of AE patients in CTD-ILD have yet to be fully elucidated.
We conducted a retrospective study of AE in CTD-ILD and IPF in our single center. The aims were to clarify the occurrence, clinical characteristics and prognosis of patients with CTD-ILD who developed AE, and to identify risk and predictive factors in Chinese population.

Methods

Study subjects

This retrospective cohort study was approved by the Ethics Committee of Nanjing Drum Tower Hospital and conducted in compliance with the Helsinki Declaration (1989) (NO.2016–160-01). We retrospectively reviewed 3280 new patients with ILD admitted to our hospital from January 2010 to December 2016, and 177 consecutive patients with an AE of underlying fILD were enrolled in our study (Fig. 1). The follow-up period was from 6 months to 7 years. The final cohort included 107 cases of AE-IPF and 70 cases of AE-CTD-ILD. All subjects had an underlying UIP or possible UIP pattern on chest high resolution computed tomography (HRCT) as defined by the American Thoracic Society (ATS)/European Respiratory Society (ERS) guidelines [1, 2]. The diagnosis of the underlying CTD and interstitial pneumonia with autoimmune features (IPAF) was according to the established criteria [2330]. IPAF means patients with ILD that had features of autoimmunity, yet falling short of a characterizable CTD, was included in CTD-ILD. The definition of AE was based on the updated international criteria for AE-IPF [3]. Briefly, AE-IPF and AE-CTD-fILD were defined as an acute, clinically significant respiratory deterioration characterized by evidence of new widespread alveolar abnormality with: ① Previous or concurrent diagnosis of IPF or a characterized CTD-fILD; ② Acute worsening or development of dyspnea typically 1 month duration; ③ CT with new bilateral ground-glass opacity and/or consolidation superimposed on a background pattern consistent with UIP or possible UIP pattern; ④ Deterioration not fully explained by cardiac failure or fluid overload [3]. All baseline and follow-up clinical data and chest imaging findings were obtained from hospital medical records. Vital status was obtained from medical records or telephone interview. Baseline clinical variables were obtained at the time of AE. The pulmonary function tests (PFTs) were conducted up to 1 month prior or after the initial diagnosis as AE-PF.

HRCT scanning

Chest HRCT examination was performed within 24 h of the diagnosis of respiratory failure with 1.0–1.5 mm thick sections with appropriate window settings (window width: 1600, window level: − 600). The findings were blindly reviewed and scored by two senior radiologists without any knowledge of clinical data. The images were assessed for the presence and extent of ground glass opacity, consolidation, traction bronchiectasis, reticulation, honeycombing and emphysema. The overall extent of abnormalities was determined for per lung using a 4-point scale (0 = no involvement, 1 = 1–25% involvement, 2 = 26–50% involvement, 3 = 51–75% involvement, and 4 = 76–100% involvement) according to the published studies [31, 32]. The HRCT of each case was evaluated as UIP pattern and possible UIP (P-UIP) pattern. Those with an inconsistent with UIP pattern were excluded from this study [1, 33].

Statistical analysis

Data are presented as mean ± standard deviation (SD) for continuous variables or percentages for categorical variables. Chi-squared and Fisher’s exact tests were used for categorical data, and the unpaired t-test and Kruskal-Wallis test were used for continuous data. A multivariate Cox regression model was used to identify significant variables capable of predicting AE or acting as prognostic factors. Bivariate correlation analysis was used to identify the relationship between total survival time and clinical variables. The incidence of AE was obtained from the Kaplan-Meier (K-M) survival curve by treating AE as the death variable. Survival was also evaluated using the log rank test. P < 0.05 was considered to represent statistical significance. Statistical analyses were performed by using IBM SPSS version 19 (SPSS, Inc., Chicago IL, USA) and Prism version 5 (GraphPad, SanDiego, CA, USA).

Results

Clinical characteristics

The baseline clinical features of subjects with AE-CTD-fILD (n = 70) and AE-IPF (n = 107) were summarized in Table 1. The average ages were similar. Female gender, prior use of corticosteroid and immunosupressants were more common in the CTD-ILD group (p<0.001, p = 0.011 and p<0.001, respectively). Serum albumin (ALB), D-dimer and TLC% pred also differed between the two groups (p<0.001, p = 0.023 and p = 0.012, respectively). Subjects with AE-CTD-ILD were treated with immunosupressants and caspofungin more frequently than AE-IPF subjects after AEs (p<0.001 and p = 0.009, respectively).
Table 1
Baseline clinical features
 
n
All patients
AE-IPF group
AE-CTD-ILD group
p value
Clinical Characteristics
 Gender (M/F)
177
116/61
81/26
35/35
< 0.001
 Age (years old)
177
67.46 ± 9.72
68.52 ± 9.87
65.84 ± 9.33
0.073
 Smoking history (Y/N)
177
56/121
38/69
18/52
0.170
 Prior corticosteroids use (Y/N)
177
125/52
68/39
57/13
0.011
 Prior immunosuppressant use (Y/N)
177
39/138
3/104
36/34
< 0.001
 Prior NAC use (Y/N)
177
153/24
91/16
62/8
0.503
 Reduction or Discontinuation of corticosteroids before AE (Y/N)
177
57/120
34/73
23/47
0.880
 Fever (Y/N)
177
105/72
62/43
43/29
0.861
 WBC count (×109)
177
11.12 ± 4.55
11.26 ± 4.91
10.91 ± 3.97
0.619
 ESR (mm/h)
177
39 (0–120)
39 (0–120)
40 (0–101)
0.275
 CRP (mg/L)
177
50.2 (0.2–246)
47 (0.2–243.5))
56.05 (0.2–246)
0.603
 LDH (U/L)
177
439.26 ± 174.13
438.28 ± 158.32
440.76 ± 197.06
0.927
 ALB (g/L)
177
32.14 ± 3.48
32.88 ± 3.45
31.01 ± 3.23
< 0.001
 BNP (pg/ml)
125
94.30 (0–1130)
104 (5–828)
88.15 (0–1130)
0.869
 PaO2/FiO2 ratio
177
126 (34–357)
121 (45–246)
140.50 (34–357)
0.050
 D-dimer (mg/L)
153
1.84 (0.02–24.82)
1.46 (0.02–22.4)
2.06 (0.10–24.82)
0.023
 CT Score
177
8 (4–8)
8 (4–8)
8 (4–8)
0.772
 FVC (L)
105
2.02 (0.74–4.61)
2.05 (0.74–3.36)
1.86 (0.86–4.61)
0.886
 FVC % pred
105
63.22 ± 18.39
60.33 ± 17.32
66.18 ± 19.20
0.170
 TLC% pred
89
53.15 ± 12.81
48.80 ± 13.53
57.67 ± 10.45
0.012
 DLCO % pred
89
41.66 ± 15.59
38.66 ± 14.78
44.78 ± 16.09
0.163
 PAH (mmHg)
89
42 ± 9.848
41.97 ± 9.642
42.03 ± 10.222
0.980
 BMI (kg/m2)
105
24.44 (17.58–34.11)
24.44 (17.99–30.53)
24.52 (17.58–34.11)
0.836
 Anti-fibrotic therapy (Y/N)
3
3/176
3/104
0/70
0.279
Treatments after AE
 Mechanical Ventilation (MV, Y/N)
177
93/84
57/50
36/34
0.810
 Maximal dosage of methylprednisonle (mg/d)
177
240 (40–1000)
240 (40–1000)
240 (40–1000)
0.252
 Immunosuppressant (Y/N)
177
58/119
14/93
44/26
< 0.001
 Immunoglobulin (Y/N)
177
72/105
43/64
29/41
0.869
 Co-trimoxazole (Y/N)
177
99/78
57/50
42/28
0.378
 Caspofungin (Y/N)
177
38/139
16/91
22/48
0.009
AE = acute exacerbation; IPF = idiopathic pulmonary fibrosis; fILD = fibrosing interstitial lung disease; CTDs = connective tissue diseases; NAC = N-acetylcysteine; WBC = white blood cell; ESR = erythrocyte sedimentation rate; CRP = C reactive protein; LDH = lactate dehydrogenase; ALB = albumin; BNP = B-type natriuretic peptide; PaO2/FiO2 = Oxygenation index; CT = Computed tomography; UIP = usual interstitial pneumonia; P-UIP = possible UIP; FVC = forced vital capacity; TLC = total lung capacity; DLCO = diffusing capacity of the lung for CO2; PAH = pulmonary arterial hypertension; BMI = body mass index; MV = mechanical ventilation

AE occurrence in patients with CTD-ILD

The incidence of AE-IPF in patients with idiopathic interstitial pneumonia (IIP) was 3.97–11.06% (6.67%) and the incidence of AE-CTD-fILD in CTD-ILD cases was 1.92–7.89% (5.99%) per year in our single center from January 2010 to December 2016 (Fig. 2A). The difference of AE occurrence between the two groups was not significant (p = 0.526) (Fig. 2B).
In univariate Cox analysis, prior corticosteroids use was the independent risk factor for AE among in subjects with CTDs after ILD diagnosis for 1 year (HR: 0.415, 95% CI: 0.215–802, p = 0.009), and pulmonary arterial hypertension (PAH) was the independent risk factor for AE occurrence in IPF patients (HR: 0.959, 95% CI: 0.921–1.000, p = 0.048) (Table 2). After considering the clinical significance and adjusting other clinical variables, prior corticosteroids and immunosuppressant use, FVC, TLC% pred, PAH and body mass index (BMI) were included in the multivariate Cox analysis. The findings showed only TLC% pred was associated with the occurrences of AE among CTD-ILD patients (HR: 0.668, 95% CI: 0.505–0.938; p:0.018). However, none of above clinical variables could predict the occurrence of AE in AE-IPF group (Table 2).
Table 2
Risk factors for AE occurrence after ILD diagnosis for 1 year by univariate and multivariate Cox regression models in the two groups
Clinical Variables
Univariate Cox Model
Multivariate Cox Model
HR
95.0%CI
p value
HR
95.0% CI
p value
AE-IPF group
 Gender
0.950
0.578–1.559
0.838
 Age (years old)
1.009
0.986–1.032
0.441
 Smoking history
1.347
0.858–2.113
0.195
0.499
0.103–2.407
0.386
 Prior corticosteroids use
0.802
0.511–1.258
0.336
0.654
0.087–4.909
0.680
 Prior immunosuppressant use
0.638
0.157–2.599
0.531
 Corticosteroids reduction or discontinuation
0.818
0.516–1.298
0.393
 FVC (L)
0.719
0.415–1.245
0.239
0.284
0.024–3.393
0.320
 TLC% pred
1.015
0.983–1.048
0.371
1.279
0.957–1.709
0.096
 DLCO% pred
0.984
0.958–1.011
0.244
 PAH (mmHg)
0.959
0.921–1.000
0.048
0.950
0.875–1.031
0.217
  BMI (kg/m2)
0.932
0.815–1.066
0.304
0.743
0.534–1.032
0.077
AE-CTD-fILD group
 Gender
0.751
0.434–1.301
0.308
 Age (years old)
0.995
0.963–1.028
0.765
 Smoking history
0.654
0.335–1.277
0.214
2.567
0.076–86.493
0.599
 Prior corticosteroids use
0.415
0.215–0.802
0.009
0.049
0.002–1.487
0.083
 Prrior immunosuppressant use
0.727
0.421–1.253
0.251
 Corticosteroids reduction or discontinuation
0.702
0.385–1.280
0.248
 FVC (L)
0.857
0.559–1.313
0.478
3.521
0.306–40.477
0.312
 TLC% pred
0.964
0.921–1.009
0.117
0.688
0.505–0.938
0.018
 DLCO% pred
1.000
0.969–1.033
0.986
 PAH (mmHg)
1.026
0.986–1.068
0.209
1.351
0.974–1.873
0.072
 BMI (kg/m2)
0.935
0.856–1.022
0.137
0.823
0.666–1.016
0.823
AE = acute exacerbation; IPF = idiopathic pulmonary fibrosis; fILD = fibrosing interstitial lung disease; CTDs = connective tissue diseases; HR = hazard ratio; 95% CI = 95% confidence interval; TLC = total lung capacity; DLCO = diffusing capacity of the lung for CO2; PAH = pulmonary arterial hypertension; BMI = body mass index

Survival

The median survival time of AE-CTD-ILD patients was 35.0 ± 4.2 days. Kaplan-Merier method analysis revealed that AE-CTD-ILD subjects had a significantly better overall survival than AE-IPF patients (log-rank test, p = 0.029) (Fig. 3A). The differences of cumulative proportion survival (CPS) less than 180 days after AE were not significant between the two groups (Additional file 1: Table S1). Among 70 cases of AE-CTD-ILD, 25 (35.7%) were primary Sjogren syndrome (PSS), 16 (22.9%) were RA, 6 (8.6%) were mixed connective tissue disease (MCTD), 5 (7.1%) were antineutrophil cytoplasmic antibodyassociated vasculitis (AAV), 8 (11.4%) were PM/DM and 10 (14.3%) were IPAF (Fig. 3B). There was no difference in survival among different CTDs subgroups (log-rank, p = 0.353) (Fig. 3C).

Prognostic factors

Bivariate correlation analysis demonstrated that overall survival time was negatively correlated with WBC counts, LDH, and CT score (r = − 0.323, p = 0.006; r = − 0.296, p = 0.013 and r = − 0.252, p = 0.035), respectively, and positively correlated to PaO2/FiO2 (r = 0.407, < 0.001) in AE-CTD-ILD group. In AE-IPF group, the findings were similar to CTD-ILD patients except for a negative correlation with the maximal dosage of corticosteroid and no correlation with PaO2/FiO2 (Table 3).
Table 3
Bivariate correlation analysis between clinical variables and overall survival time
Clinical Variables
AE-IPF group
AE-CTD-fILD group
r
p value
r
p value
 WBC count (× 109)
−0.277
0.004
−0.323
0.006
 LDH (U/L)
−0.220
0.023
−0.296
0.013
 PaO2/FiO2 ratio
0.158
0.104
0.407
< 0.001
 CT score
−0.433
< 0.001
−0.252
0.035
 Maximal dosage of methylprednisolone (mg/d)
−0.196
0.043
−0.146
0.227
WBC: white blood cell; LDH: lactate dehydrogenase; PaO2/FiO2 ratio: Oxygenation index; HRCT: high resolution computed tomography; MV: mechanical ventilation
The univariate analysis showed that WBC count, CRP, LDH, ALB, D-dimmer, PaO2/FiO2, CT score, treatment with MV, maximal dosage of methylprednisolone, immunosuppressant, immunoglobulin, co-trimoxazole and caspofungin for AE were associated with survival in all 177 cases (all p<0.05, respectively), while only WBC count, PaO2/FiO2 and CT score were the independent risk factors for survival in these patients (HR 1.051, 95%CI 1.013–1.090, p = 0.001; HR 0.991, 95%CI 0.987–0.994, p<0.001 and HR 1.337, 95%CI 1.063–1.682, p<0.013, respectively) (Table 4). As shown in Table 5, similar to all 177 patients, WBC count, PaO2/FiO2 ratio and CT score in AE-IPF group were also the independent risk factors for survival by a multivariate Cox regression analysis (HR 1.067, 95%CI 1.028–1.108, p = 0.001; HR 0.993, 95%CI 0.989–0.998, p = 0.002 and HR 1.531, 95%CI 1.170–2.002, p = 0.002, respectively).
Table 4
Prognostic factors for survival by univariate and multivariate Cox regression models in all patients
Clinical Variables
Univariate Cox Model
Multivariate Cox Model
HR
95.0% CI
p value
HR
95.0% CI
p value
 WBC count (×109)
1.075
1.041–1.109
<0.001
1.051
1.013–1.090
0.008
 CRP (mg/L)
1.003
1.000–1.006
0.038
1.003
0.999–1.007
0.157
 LDH (U/L)
1.003
1.002–1.004
<0.001
1.000
0.998–1.001
0.430
 ALB (U/L)
0.943
0.987–0.992
0.022
1.000
0.999–1.001
0.545
 D-dimmer
1.048
1.018–1.080
0.002
1.021
0.971–1.074
0.421
 PaO2/FiO2 ratio
0.991
0.998–0.994
<0.001
0.991
0.987–0.994
<0.001
 CT score
1.402
1.195–1.645
<0.001
1.337
1.063–1.682
0.013
 MV
2.877
2.070–4.000
<0.001
 Maximal dosage of methylprednisolone (mg/d)
1.001
1.000–1.001
0.016
1.000
0.999–1.001
0.840
 Immunosuppressant
0.673
0.478–0.947
0.023
 Immunoglobulin
2.103
1.524–2.901
<0.001
 Co-trimoxazole
1.382
1.009–1.892
0.044
 Caspofungin
2.057
1.406–3.009
<0.001
WBC: white blood cell; LDH: lactate dehydrogenase; PaO2/FiO2ratio: Oxygenation index; MV: mechanical ventilation; HRCT: high resolution computed tomography
Table 5
Prognostic factors for survival by C by univariate and multivariate Cox regression models in the two groups
Clinical Variables
Univariate Cox Model
Multivariate Cox Model
HR
95.0% CI
p value
HR
95.0% CI
p value
AE-IPF group
 WBC count (×109)
1.071
1.033–1.111
< 0.001
1.069
1.031–1.109
< 0.001
 LDH (U/L)
1.002
1.001–1.003
0.003
 PaO2/FiO2 ratio
0.993
0.989–0.997
0.001
0.993
0.989–0.997
0.002
 CT score
1.703
1.343–2.158
< 0.001
1.573
1.211–2.042
0.001
 MV
2.673
1.747–4.087
< 0.001
 Maximal dosage of methylprednisolone (mg/d)
1.001
1.000–1.002
0.006
1.000
1.000–1.001
0.534
 Immunoglobulin
2.182
1.427–3.338
< 0.001
 Caspofungin
1.809
1.038–3.153
0.036
AE-CTD-fILD group
 WBC count (×109)
1.098
1.027–1.175
0.006
1.074
1.004–1.150
0.038
 LDH (U/L)
1.004
1.002–1.005
< 0.001
   
 PaO2/FiO2 ratio
0.989
0.985–0.994
< 0.001
0.989
0.984–0.994
< 0.001
 CT score
1.253
0.993–1.582
0.058
0.975
0.746–1.274
0.852
 MV
3.495
2.020–6.047
< 0.001
   
 Maximal dosage of methylprednisolone (mg/d)
1.000
0.999–1.001
0.613
0.999
0.998–1.000
0.277
 Immunoglobulin
2.370
1.401–4.009
0.001
 Caspofungin
2.741
1.563–4.808
< 0.001
WBC: white blood cell; LDH: lactate dehydrogenase; PaO2/FiO2 ratio: Oxygenation index; MV: mechanical ventilation; HRCT: high resolution computed tomography
In AE-CTD-ILD group, WBC counts, LDH, PaO2/FiO2 ratio, treatment with MV, immunoglobulin, and caspofungin for AE were associated with the survival by univariate Cox model (all p<0.05, respectively). After considering the clinical significance and adjusting other clinical variables, WBC count, PaO2/FiO2, CT score and maximal dosage of methylprednisolone were included in multivariate Cox model. The findings showed that WBC count and PaO2/FiO2 ratio were the independent prognostic factors for survival by a multivariate Cox regression analysis (HR 1.074, 95%CI 1.004–1.150, p = 0.038 and HR 0.989, 95%CI 0.984–0.994, p<0.001, respectively) (Table 5).

Discussion

The present study retrospectively compared the clinical, chest imaging and follow-up data between 70 subjects with AE-CTD-ILD and 107 subjects with AE-IPF. The clinical features of patients with AE-CTD-ILD differed from those of AE-IPF. Subjects with AE-CTD-ILD had a better survival than AE-IPF patients, while the survival of AE cases in the CTD subgroups didn’t show any difference. WBC count and PO2/FiO2 ratio could predict the survival of AE-CTD-ILD patients independently.
Recently, the definition and diagnostic criteria of AE-IPF have been updated. IPF patients manifesting an acute, clinically significant respiratory deterioration characterized by evidence of new widespread alveolar abnormality on chest imaging or histopathology should be considered as having an AE [3]. Data showed that a potential infectious aetiology is up to one-third of patients with ILD and acute respiratory decline [22]. The updated criteria of AE-IPF do not exclude respiratory infection [3]. The causes of acute lung injure (ALI) including infection, aspiration, drugs and surgery may lead to events that are indistinguishable from idiopathic AE of IPF [3]. These events are further classified into “triggered” (ie, postprocedure, drug toxicity, infection, aspiration) or “idiopathic” (ie, no inciting event identified) AEs in the updated guideline [3].
The published reports about AE in patients with CTD-ILD have been relatively small series and show inconsistent results. The incidence of AE in RA-UIP was about 11.1–20% [18, 19]. The mild elevation of mean PAH was very common in CTD-ILD patients [34]. Suda et al. suggested that the clinical characteristics of AE in CTD-ILD were similar to those of IPF with high mortality [19]. Park and Suda reported that AE occurred mostly with RA-UIP with poor outcome [18, 19]. Song and Huie suggested that CTD-UIP group was younger, included more women and nonsmokers, and showed better survival than the IPF-UIP group [7, 22]. The current study was based on the updated criteria of Collard about AE-IPF [3] and comprised a 177 cases of AE patients with IPF and CTD-ILD. The findings showed that the clinical features, treatment and prognosis of patients with AE-CTD-ILD differed from those with AE-IPF and published reports [7, 8, 18, 35].
The incidence of AE in IPF patients has been reported in a number of studies. Due to different definition and diagnostic criteria of AE, AE incidence in IPF varied greatly, estimated to be in the range of 4–20% per year [48]. In patients with CTD-ILD, Suda et al. revealed an overall AE incidence of 7.2% and a 1-year incidence of 1.25% [19]. Compared with AE-IPF, the incidence of AE in CTD-ILD seemed to be lower [19]. In our single center, AE incidence was about 5% in all new ILD patients about 6% per year in those with CTD-ILD. AE incidence of fILD patients in our study is similar to the published reports of IPF population in other countries [48]. IPF patients with physiologically and functionally advanced disease are thought to be at higher risk of AE [3]. However, little is known about the causative factors of AE in patients with CTD-ILD. In CTD-ILD patients, age and SS overlapped with PM/DM were the independent risk factors for AE [19, 36]. In our study, AE occurrence in CTD-ILD patients within 1 year after ILD diagnosis was weakly associated with TLC% pred, likely due to the limited sample size. A larger sample prospective study of AE-CTD-ILD is therefore needed to clarify this complication.
The poor survival of patients with AE-IPF is a great challenge. The MST of these patients was about 3–4 months [6, 7]. Nearly half of patients with AE-IPF died in the hospital, and patients with both definite AE-IPF and suspected AE-IPF had a similar clinical outcome [6, 7, 22]. The published reports on AE-CTD-ILD differ from AE-IPF and include a relatively small number of cases. Tachikawa et al. concluded that the survival of AE-CTD-ILD was better than that of advanced IPF (90-day mortality at 33% vs. 69%) [37]. Huie et al. suggested that IPF patients had a lower rate of survival as compared with patients with non-IPF fibrotic disease after acute respiratory decline [22]. Suda et al. suggested that AE incidence in CTD-ILD was similar to that of IPF with poor prognosis [19]. In our cohort study, the overall survival was significant better in subjects with AE-CTD-ILD than AE-IPF cases, despite their similar short-term survival. Usui Y.et al. reported that systemic inflammatory response (SIRS) was the most significant predictor for in-hospital mortality in patients with AE of chronic fILD including IPF and non-IPF [38]. Infection at AE was thought to determine the short-term survival in these patients. In current study, the inflammatory indicators, WBC counts, CRP, LDH and CT scores didn’t show any difference between the two groups, implying that infection was similar between the two groups. So, the similar short-term survival may be associated with the similar infection in the two groups [38]. The better overall survival in CTD-ILD patients may be associated with the favorable response to corticosteroids treatment [23]. Yoshimura et al. also suggested that the diagnosis of IPF might predict a favorable prognosis in patients with chronic fILD [39], thereby a better survival in AE-CTD-fILD. The lack of difference in overall survival among different CTD subgroups may be due to the small sample size in our study.
A number of factors including lower baseline FVC and DLCO, extensive CT abnormalities and CT pattern, worse oxygenation, higher bronchoalveolar lavage neutrophil and lymphocyte percentages at the time of AE were reported to be associated with the survival of patients with AE-IPF [7, 8, 15, 32, 40, 41]. Literature also indicated that described age, TLC, honeycombing score and higher initial MPAP were the independent prognostic factors in AE-CTD-ILD [34, 35]. In current study, WBC count and PO2/FiO2 ratio could predict the survival of patients with AE-CTD-ILD after adjusting for other clinical variables. Now, infection has been regarded as one of the causes for AE in IPF patients [3]. WBC is a non-specific inflammation marker of infection. Infection with elevated WBC counts was correlated with the poor clinical outcomes in AE-CTD-fILD patients in our study. Low PO2/FiO2 ratio could account for the severity of hypoxia and clinical condition at AE, which indicated patients with more severe condition when AE occurred would have worse prognosis. The different risk factors of prognosis in the two groups may be due to the underlying diseases and the conditions at AE occurrence.
Although there is no effective therapy for AE-IPF, supportive care including supplemental oxygen and palliation of symptoms are regarded as important treatment modality. The use of MV is controversial [1, 3]. Corticosteroids are recommended but with somewhat low-quality evidence [1, 3]. There is no consensus on the dosage and course of therapy. A slight survival benefit was reported for the treatment of AE-IPF combining cyclosporine with systemic glucocorticoids as compared to that without cyclosporine in a small retrospective study [41]. The therapy of steroids combined with cyclophosphosphamide for patients with AE-CTD-ILD appeared better than those without any immunosuppressant [7, 42]. However, there is no evidence supporting the benefit of a specific immunosuppressant. Addition of intravenous thrombomodulin and polymyxin B immobilized fiber column perfusion to conventional treatment was shown to improve the survival in patients with AE-IPF [39, 41]. Rituximab combined with plasma exchange and intravenous immunoglobulin also showed certain benefits [42]. All patients in our study used corticosteroids after AEs, but the maximal dosage was different for each case according to the clinical severity and the extent of damage on chest CT. The overall survival time was not correlated with the maximal dosage of methylprednisolone both in CTD-ILD and IPF patients when AE occurred. So, we speculated that the higher dosage of corticosteroids would not be beneficial for AE patients, and the clinical outcomes of AE patients with pulmonary fibrosis mainly depend on the underlying clinical conditions and the extent of lung injury at AE.
The current study has some limitations. It was designed retrospectively and the stable cases of pulmonary fibrosis were not available as controls. Histological evidence was not available for the diagnosis of pulmonary fibrosis. It was unable to get the exact number of cases with IPF patients. A definite cause of death was not available for each case, although most patients died of AE. A prospective, multicenter, multinational study of a larger cases cohort would be helpful to provide enough data for the epidemiology in AE-CTD-ILD.

Conclusions

In summary, the current study demonstrated that the clinical characteristics of patients with AE-CTD-ILD differed from those of AE-IPF. The incidence of AE-CTD-ILD was similar to that of AE-IPF. Subjects with AE-CTD-fILD had a better prognosis than those with AE-IPF. However, the survival of AE patients in each group was identical. WBC count and PO2/FiO2 were the independent predictors for the survival in patients with CTD-fILD. A higher dosage of corticosteroids would not be beneficial for the survival of fILD patien.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12890-019-0960-1.

Acknowledgements

We would like to thank the subjects and their families who participated in this study and all people who participated in the clinical management. We also would like to thank Prof. Kevin K, Brown (Department of medicine, National Jewish Health, USA) and Prof. Yin Chen (Department of Pharmacology, University of Arizona, Tucson, USA) for reviewing and editing this manuscript. The authors were fully responsible for all content and editorial decisions, were involved at all stages of development and provided their approval on the final version.
This study was approved by the Ethics Committee of Nanjing Drum Tower Hospital (NO.2016–160-01) in compliance with the Helsinki Declaration along with established written informed consent. This research was carried out in accordance with the approved guidelines. Written informed consent was obtained from all participants before inclusion.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. American Journal of Respiratory & Critical Care Medicine. 2011;183(6):788–824.CrossRef Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. American Journal of Respiratory & Critical Care Medicine. 2011;183(6):788–824.CrossRef
2.
Zurück zum Zitat Travis WD, Costabel U, Hansell DM, King TE Jr, Lynch DA, Nicholson AG, Ryerson CJ, Ryu JH, Selman M, Wells AU, et al. An official American Thoracic Society/European Respiratory Society statement: update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188(6):733–48.CrossRef Travis WD, Costabel U, Hansell DM, King TE Jr, Lynch DA, Nicholson AG, Ryerson CJ, Ryu JH, Selman M, Wells AU, et al. An official American Thoracic Society/European Respiratory Society statement: update of the international multidisciplinary classification of the idiopathic interstitial pneumonias. Am J Respir Crit Care Med. 2013;188(6):733–48.CrossRef
3.
Zurück zum Zitat Collard HR, Ryerson CJ, Corte TJ, Jenkins G, Kondoh Y, Lederer DJ, Lee JS, Maher TM, Wells AU, Antoniou KM. Acute Exacerbation of Idiopathic Pulmonary Fibrosis. An International Working Group Report. American Journal of Respiratory & Critical Care Medicine. 2016;194(3). Collard HR, Ryerson CJ, Corte TJ, Jenkins G, Kondoh Y, Lederer DJ, Lee JS, Maher TM, Wells AU, Antoniou KM. Acute Exacerbation of Idiopathic Pulmonary Fibrosis. An International Working Group Report. American Journal of Respiratory & Critical Care Medicine. 2016;194(3).
4.
Zurück zum Zitat Ryerson CJ, Cottin V, Brown KK, Collard HR. Acute exacerbation of idiopathic pulmonary fibrosis: shifting the paradigm. Eur Respir J. 2015;46(2):512–20.CrossRef Ryerson CJ, Cottin V, Brown KK, Collard HR. Acute exacerbation of idiopathic pulmonary fibrosis: shifting the paradigm. Eur Respir J. 2015;46(2):512–20.CrossRef
5.
Zurück zum Zitat Atkins CP, Loke YK, Wilson AM. S16 outcomes in idiopathic pulmonary fibrosis: a meta-analysis from placebo controlled trials. Respir Med. 2014;108(2):376–87.CrossRef Atkins CP, Loke YK, Wilson AM. S16 outcomes in idiopathic pulmonary fibrosis: a meta-analysis from placebo controlled trials. Respir Med. 2014;108(2):376–87.CrossRef
6.
Zurück zum Zitat Collard HR, Yow E, Richeldi L, Anstrom KJ, Glazer C: Suspected acute exacerbation of idiopathic pulmonary fibrosis as an outcome measure in clinical trials. Respiratory Research,14,1(2013-07-13) 2013, 14(1):1–7. Collard HR, Yow E, Richeldi L, Anstrom KJ, Glazer C: Suspected acute exacerbation of idiopathic pulmonary fibrosis as an outcome measure in clinical trials. Respiratory Research,14,1(2013-07-13) 2013, 14(1):1–7.
7.
Zurück zum Zitat Song JW, Hong SB, Lim CM, Koh Y, Kim DS. Acute exacerbation of idiopathic pulmonary fibrosis: incidence, risk factors and outcome. Eur Respir J. 2011;37(2):356.CrossRef Song JW, Hong SB, Lim CM, Koh Y, Kim DS. Acute exacerbation of idiopathic pulmonary fibrosis: incidence, risk factors and outcome. Eur Respir J. 2011;37(2):356.CrossRef
8.
Zurück zum Zitat Simon-Blancal V, Freynet O, Nunes H, Bouvry D, Naggara N, Brillet PY, Denis D, Cohen Y, Vincent F, Valeyre D, et al. Acute exacerbation of idiopathic pulmonary fibrosis: outcome and prognostic factors. Respiration; international review of thoracic diseases. 2012;83(1):28–35.CrossRef Simon-Blancal V, Freynet O, Nunes H, Bouvry D, Naggara N, Brillet PY, Denis D, Cohen Y, Vincent F, Valeyre D, et al. Acute exacerbation of idiopathic pulmonary fibrosis: outcome and prognostic factors. Respiration; international review of thoracic diseases. 2012;83(1):28–35.CrossRef
9.
Zurück zum Zitat Natsuizaka M, Chiba H, Kuronuma K, Otsuka M, Kudo K, Mori M, Bando M, Sugiyama Y, Takahashi H. Epidemiologic survey of Japanese patients with idiopathic pulmonary fibrosis and investigation of ethnic differences. Am J Respir Crit Care Med. 2014;190(7):773–9.CrossRef Natsuizaka M, Chiba H, Kuronuma K, Otsuka M, Kudo K, Mori M, Bando M, Sugiyama Y, Takahashi H. Epidemiologic survey of Japanese patients with idiopathic pulmonary fibrosis and investigation of ethnic differences. Am J Respir Crit Care Med. 2014;190(7):773–9.CrossRef
10.
Zurück zum Zitat Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE Jr, Lasky JA, Loyd JE, Noth I, Olman MA, et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2007;176(7):636–43.CrossRef Collard HR, Moore BB, Flaherty KR, Brown KK, Kaner RJ, King TE Jr, Lasky JA, Loyd JE, Noth I, Olman MA, et al. Acute exacerbations of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2007;176(7):636–43.CrossRef
11.
Zurück zum Zitat Johannson KA, Vittinghoff E, Lee K, Balmes JR, Ji W, Kaplan GG, Kim DS, Collard HR. Acute exacerbation of idiopathic pulmonary fibrosis associated with air pollution exposure. Eur Respir J. 2014;43(4):1124–31.CrossRef Johannson KA, Vittinghoff E, Lee K, Balmes JR, Ji W, Kaplan GG, Kim DS, Collard HR. Acute exacerbation of idiopathic pulmonary fibrosis associated with air pollution exposure. Eur Respir J. 2014;43(4):1124–31.CrossRef
12.
Zurück zum Zitat Mura M, Porretta MA, Bargagli E, Sergiacomi G, Zompatori M, Sverzellati N, Taglieri A, Mezzasalma F, Rottoli P, Saltini C, et al. Predicting survival in newly diagnosed idiopathic pulmonary fibrosis: a 3-year prospective study. Eur Respir J. 2012;40(1):101–9.CrossRef Mura M, Porretta MA, Bargagli E, Sergiacomi G, Zompatori M, Sverzellati N, Taglieri A, Mezzasalma F, Rottoli P, Saltini C, et al. Predicting survival in newly diagnosed idiopathic pulmonary fibrosis: a 3-year prospective study. Eur Respir J. 2012;40(1):101–9.CrossRef
13.
Zurück zum Zitat Kondoh Y, Taniguchi H, Ebina M, Azuma A, Ogura T, Taguchi Y, Suga M, Takahashi H, Nakata K, Sugiyama Y, et al. Risk factors for acute exacerbation of idiopathic pulmonary fibrosis--extended analysis of pirfenidone trial in Japan. Respir Investig. 2015;53(6):271–8.CrossRef Kondoh Y, Taniguchi H, Ebina M, Azuma A, Ogura T, Taguchi Y, Suga M, Takahashi H, Nakata K, Sugiyama Y, et al. Risk factors for acute exacerbation of idiopathic pulmonary fibrosis--extended analysis of pirfenidone trial in Japan. Respir Investig. 2015;53(6):271–8.CrossRef
14.
Zurück zum Zitat Reichmann WM, Yu YF, Macaulay D, Wu EQ, Nathan SD. Change in forced vital capacity and associated subsequent outcomes in patients with newly diagnosed idiopathic pulmonary fibrosis. Bmc Pulmonary Medicine. 2015;15(1):167.CrossRef Reichmann WM, Yu YF, Macaulay D, Wu EQ, Nathan SD. Change in forced vital capacity and associated subsequent outcomes in patients with newly diagnosed idiopathic pulmonary fibrosis. Bmc Pulmonary Medicine. 2015;15(1):167.CrossRef
15.
Zurück zum Zitat Kim DS, Park JH, Park BK, Lee JS, Nicholson AG, Colby T. Acute exacerbation of idiopathic pulmonary fibrosis: frequency and clinical features. Eur Respir J. 2006;27(1):143–50.CrossRef Kim DS, Park JH, Park BK, Lee JS, Nicholson AG, Colby T. Acute exacerbation of idiopathic pulmonary fibrosis: frequency and clinical features. Eur Respir J. 2006;27(1):143–50.CrossRef
16.
Zurück zum Zitat Ohshimo S, Ishikawa N, Horimasu Y, Hattori N, Hirohashi N, Tanigawa K, Kohno N, Bonella F, Guzman J, Costabel U. Baseline KL-6 predicts increased risk for acute exacerbation of idiopathic pulmonary fibrosis. Respir Med. 2014;108(7):1031–9.CrossRef Ohshimo S, Ishikawa N, Horimasu Y, Hattori N, Hirohashi N, Tanigawa K, Kohno N, Bonella F, Guzman J, Costabel U. Baseline KL-6 predicts increased risk for acute exacerbation of idiopathic pulmonary fibrosis. Respir Med. 2014;108(7):1031–9.CrossRef
17.
Zurück zum Zitat Cao M, Swigris JJ, Xin W, Min C, Qiu Y, Mei H, Xiao Y, Cai H. Plasma Leptin is elevated in acute exacerbation of idiopathic pulmonary fibrosis. Mediat Inflamm. 2016;2016(7):1–7.CrossRef Cao M, Swigris JJ, Xin W, Min C, Qiu Y, Mei H, Xiao Y, Cai H. Plasma Leptin is elevated in acute exacerbation of idiopathic pulmonary fibrosis. Mediat Inflamm. 2016;2016(7):1–7.CrossRef
18.
Zurück zum Zitat Park IN, Dong SK, Shim TS, Lim CM, Sang DL, Koh Y, Kim WS, Kim WD, Jang SJ, Colby TV. Acute exacerbation of interstitial pneumonia other than idiopathic pulmonary fibrosis. Chest. 2007;132(1):214–20.CrossRef Park IN, Dong SK, Shim TS, Lim CM, Sang DL, Koh Y, Kim WS, Kim WD, Jang SJ, Colby TV. Acute exacerbation of interstitial pneumonia other than idiopathic pulmonary fibrosis. Chest. 2007;132(1):214–20.CrossRef
19.
Zurück zum Zitat Suda T, Kaida Y, Nakamura Y, Enomoto N, Fujisawa T, Imokawa S, Hashizume H, Naito T, Dai H, Takehara Y. Acute exacerbation of interstitial pneumonia associated with collagen vascular diseases. Respir Med. 2009;103(6):846–53.CrossRef Suda T, Kaida Y, Nakamura Y, Enomoto N, Fujisawa T, Imokawa S, Hashizume H, Naito T, Dai H, Takehara Y. Acute exacerbation of interstitial pneumonia associated with collagen vascular diseases. Respir Med. 2009;103(6):846–53.CrossRef
20.
Zurück zum Zitat Miyazaki Y, Tateishi T, Akashi T, Ohtani Y, Inase N, Yoshizawa Y. Clinical predictors and histologic appearance of acute exacerbations in chronic hypersensitivity pneumonitis. Chest. 2008;134(6):1265.CrossRef Miyazaki Y, Tateishi T, Akashi T, Ohtani Y, Inase N, Yoshizawa Y. Clinical predictors and histologic appearance of acute exacerbations in chronic hypersensitivity pneumonitis. Chest. 2008;134(6):1265.CrossRef
21.
Zurück zum Zitat Olson AL, Huie TJ, Groshong SD, Cosgrove GP, Janssen WJ, Schwarz MI, Brown KK, Frankel SK. Acute exacerbations of fibrotic hypersensitivity pneumonitis: a case series. Chest. 2008;134(4):844–50.CrossRef Olson AL, Huie TJ, Groshong SD, Cosgrove GP, Janssen WJ, Schwarz MI, Brown KK, Frankel SK. Acute exacerbations of fibrotic hypersensitivity pneumonitis: a case series. Chest. 2008;134(4):844–50.CrossRef
22.
Zurück zum Zitat Huie TJ, Olson AL, Cosgrove GP, Janssen WJ, Lara AR, Lynch DA, Groshong SD, Moss M, Schwarz MI, Brown KK. A detailed evaluation of acute respiratory decline in patients with fibrotic lung disease: Aetiology and outcomes. Respirology. 2010;15(6):909–17.CrossRef Huie TJ, Olson AL, Cosgrove GP, Janssen WJ, Lara AR, Lynch DA, Groshong SD, Moss M, Schwarz MI, Brown KK. A detailed evaluation of acute respiratory decline in patients with fibrotic lung disease: Aetiology and outcomes. Respirology. 2010;15(6):909–17.CrossRef
23.
Zurück zum Zitat Toyoda Y, Hanibuchi M, Kishi J, Kawano H, Morizumi S, Sato S, Kondo M, Takikura T, Tezuka T, Goto H. Clinical features and outcome of acute exacerbation of interstitial pneumonia associated with connective tissue disease. Journal of Medical Investigation. 2016;63(3–4):294.CrossRef Toyoda Y, Hanibuchi M, Kishi J, Kawano H, Morizumi S, Sato S, Kondo M, Takikura T, Tezuka T, Goto H. Clinical features and outcome of acute exacerbation of interstitial pneumonia associated with connective tissue disease. Journal of Medical Investigation. 2016;63(3–4):294.CrossRef
24.
Zurück zum Zitat Arnett FC, Edworthy SM, Bloch DA, DJ MS, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS. The American rheumatism association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31(3):315–24.CrossRef Arnett FC, Edworthy SM, Bloch DA, DJ MS, Fries JF, Cooper NS, Healey LA, Kaplan SR, Liang MH, Luthra HS. The American rheumatism association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31(3):315–24.CrossRef
25.
Zurück zum Zitat Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, et al. Rheumatoid arthritis classification criteria: an American College of Rheumatology/European league against rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–81.CrossRef Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, et al. Rheumatoid arthritis classification criteria: an American College of Rheumatology/European league against rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–81.CrossRef
26.
Zurück zum Zitat Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975;292(7):344–7.CrossRef Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts). N Engl J Med. 1975;292(7):344–7.CrossRef
27.
Zurück zum Zitat Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982;25(11):1271–7.CrossRef Tan EM, Cohen AS, Fries JF, Masi AT, McShane DJ, Rothfield NF, Schaller JG, Talal N, Winchester RJ. The 1982 revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1982;25(11):1271–7.CrossRef
28.
Zurück zum Zitat Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.CrossRef Hochberg MC. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40(9):1725.CrossRef
29.
Zurück zum Zitat Fujibayashi T, Sugai S, Miyasaka N, Hayashi Y, Tsubota K. Revised Japanese criteria for Sjogren's syndrome (1999): availability and validity. Mod Rheumatol. 2004;14(6):425–34.CrossRef Fujibayashi T, Sugai S, Miyasaka N, Hayashi Y, Tsubota K. Revised Japanese criteria for Sjogren's syndrome (1999): availability and validity. Mod Rheumatol. 2004;14(6):425–34.CrossRef
30.
Zurück zum Zitat Fischer A, Antoniou KM, Brown KK, Cadranel J, Corte TJ, du Bois RM, Lee JS, Leslie KO, Lynch DA, Matteson EL, et al. An official European Respiratory Society/American Thoracic Society research statement: interstitial pneumonia with autoimmune features. Eur Respir J. 2015;46(4):976–87.CrossRef Fischer A, Antoniou KM, Brown KK, Cadranel J, Corte TJ, du Bois RM, Lee JS, Leslie KO, Lynch DA, Matteson EL, et al. An official European Respiratory Society/American Thoracic Society research statement: interstitial pneumonia with autoimmune features. Eur Respir J. 2015;46(4):976–87.CrossRef
31.
Zurück zum Zitat Lynch DA, Godwin JD, Safrin S, Starko KM, Hormel P, Brown KK, Raghu G, King TE Jr, Bradford WZ, Schwartz DA, et al. High-resolution computed tomography in idiopathic pulmonary fibrosis: diagnosis and prognosis. Am J Respir Crit Care Med. 2005;172(4):488–93.CrossRef Lynch DA, Godwin JD, Safrin S, Starko KM, Hormel P, Brown KK, Raghu G, King TE Jr, Bradford WZ, Schwartz DA, et al. High-resolution computed tomography in idiopathic pulmonary fibrosis: diagnosis and prognosis. Am J Respir Crit Care Med. 2005;172(4):488–93.CrossRef
32.
Zurück zum Zitat Akira M, Kozuka T, Yamamoto S, Sakatani M. Computed tomography findings in acute exacerbation of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;178(4):372–8.CrossRef Akira M, Kozuka T, Yamamoto S, Sakatani M. Computed tomography findings in acute exacerbation of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2008;178(4):372–8.CrossRef
33.
Zurück zum Zitat Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, Behr J, Cottin V, Danoff SK, Morell F, Flaherty KR, Wells A, Martinez FJ, Azuma A, Bice TJ, Bouros D, Brown KK, Collard HR, Duggal A, Galvin L, Inoue Y, Jenkins RG, Johkoh T, Kazerooni EA, Kitaichi M, Knight SL, Mansour G, Nicholson AG, Pipavath SNJ, Buendía-Roldán I, Selman M, Travis WD, Walsh S, Wilson KC; American Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin American Thoracic Society. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med. 2018;198(5):e44–68. Raghu G, Remy-Jardin M, Myers JL, Richeldi L, Ryerson CJ, Lederer DJ, Behr J, Cottin V, Danoff SK, Morell F, Flaherty KR, Wells A, Martinez FJ, Azuma A, Bice TJ, Bouros D, Brown KK, Collard HR, Duggal A, Galvin L, Inoue Y, Jenkins RG, Johkoh T, Kazerooni EA, Kitaichi M, Knight SL, Mansour G, Nicholson AG, Pipavath SNJ, Buendía-Roldán I, Selman M, Travis WD, Walsh S, Wilson KC; American Thoracic Society, European Respiratory Society, Japanese Respiratory Society, and Latin American Thoracic Society. Diagnosis of Idiopathic Pulmonary Fibrosis. An Official ATS/ERS/JRS/ALAT Clinical Practice Guideline. Am J Respir Crit Care Med. 2018;198(5):e44–68.
34.
Zurück zum Zitat Takahashi K, Taniguchi H, Ando M, Sakamoto K, Kondoh Y, Watanabe N, Kimura T, Kataoka K, Suzuki A, Ito S, et al. Mean pulmonary arterial pressure as a prognostic indicator in connective tissue disease associated with interstitial lung disease: a retrospective cohort study. BMC Pulm Med. 2016;16(1):55.CrossRef Takahashi K, Taniguchi H, Ando M, Sakamoto K, Kondoh Y, Watanabe N, Kimura T, Kataoka K, Suzuki A, Ito S, et al. Mean pulmonary arterial pressure as a prognostic indicator in connective tissue disease associated with interstitial lung disease: a retrospective cohort study. BMC Pulm Med. 2016;16(1):55.CrossRef
35.
Zurück zum Zitat Jin WS, Do KH, Kim MY, Jang SJ, Colby TV, Dong SK. Pathologic and radiologic differences between idiopathic and collagen vascular disease-related usual interstitial pneumonia. Chest. 2009;136(1):23–30.CrossRef Jin WS, Do KH, Kim MY, Jang SJ, Colby TV, Dong SK. Pathologic and radiologic differences between idiopathic and collagen vascular disease-related usual interstitial pneumonia. Chest. 2009;136(1):23–30.CrossRef
36.
Zurück zum Zitat Tomiyama F, Watanabe R, Ishii T, Kamogawa Y, Fujita Y, Shirota Y, Sugimura K, Fujii H, Harigae H. High prevalence of acute exacerbation of interstitial lung disease in Japanese patients with systemic sclerosis. Tohoku J Exp Med. 2016;239(4):297.CrossRef Tomiyama F, Watanabe R, Ishii T, Kamogawa Y, Fujita Y, Shirota Y, Sugimura K, Fujii H, Harigae H. High prevalence of acute exacerbation of interstitial lung disease in Japanese patients with systemic sclerosis. Tohoku J Exp Med. 2016;239(4):297.CrossRef
37.
Zurück zum Zitat Tachikawa R, Tomii K, Ueda H, Nagata K, Nanjo S, Sakurai A, Otsuka K, Kaji R, Hayashi M, Katakami N, et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia: collagen vascular diseases-related versus idiopathic. Respiration; international review of thoracic diseases. 2012;83(1):20–7.CrossRef Tachikawa R, Tomii K, Ueda H, Nagata K, Nanjo S, Sakurai A, Otsuka K, Kaji R, Hayashi M, Katakami N, et al. Clinical features and outcome of acute exacerbation of interstitial pneumonia: collagen vascular diseases-related versus idiopathic. Respiration; international review of thoracic diseases. 2012;83(1):20–7.CrossRef
38.
Zurück zum Zitat Usui Y, Kaga A, Sakai F, Shiono A, Komiyama K, Hagiwara K, Kanazawa M. A cohort study of mortality predictors in patients with acute exacerbation of chronic fibrosing interstitial pneumonia. BMJ Open. 2013:3(7). Usui Y, Kaga A, Sakai F, Shiono A, Komiyama K, Hagiwara K, Kanazawa M. A cohort study of mortality predictors in patients with acute exacerbation of chronic fibrosing interstitial pneumonia. BMJ Open. 2013:3(7).
39.
Zurück zum Zitat Abe S, Azuma A, Mukae H, Ogura T, Taniguchi H, Bando M, Sugiyama Y. Polymyxin B-immobilized fiber column (PMX) treatment for idiopathic pulmonary fibrosis with acute exacerbation: a multicenter retrospective analysis. Intern Med. 2012;51(12):1487–91.CrossRef Abe S, Azuma A, Mukae H, Ogura T, Taniguchi H, Bando M, Sugiyama Y. Polymyxin B-immobilized fiber column (PMX) treatment for idiopathic pulmonary fibrosis with acute exacerbation: a multicenter retrospective analysis. Intern Med. 2012;51(12):1487–91.CrossRef
40.
Zurück zum Zitat Sakamoto S, Homma S, Miyamoto A, Kurosaki A, Fujii T, Yoshimura K. Cyclosporin a in the treatment of acute exacerbation of idiopathic pulmonary fibrosis. Intern Med. 2010;49(2):109–15.CrossRef Sakamoto S, Homma S, Miyamoto A, Kurosaki A, Fujii T, Yoshimura K. Cyclosporin a in the treatment of acute exacerbation of idiopathic pulmonary fibrosis. Intern Med. 2010;49(2):109–15.CrossRef
41.
Zurück zum Zitat Kataoka K, Taniguchi H, Kondoh Y, Nishiyama O, Kimura T, Matsuda T, Yokoyama T, Sakamoto K, Ando M. Recombinant human Thrombomodulin in acute exacerbation of idiopathic pulmonary fibrosis. Chest. 2015;148(2):436–43.CrossRef Kataoka K, Taniguchi H, Kondoh Y, Nishiyama O, Kimura T, Matsuda T, Yokoyama T, Sakamoto K, Ando M. Recombinant human Thrombomodulin in acute exacerbation of idiopathic pulmonary fibrosis. Chest. 2015;148(2):436–43.CrossRef
42.
Zurück zum Zitat Donahoe M, Valentine VG, Chien N, Gibson KF, Raval JS, Saul M, Xue J, Zhang Y, Duncan SR. Autoantibody-targeted treatments for acute exacerbations of idiopathic pulmonary fibrosis. PLoS One. 2015;10(6):e0127771.CrossRef Donahoe M, Valentine VG, Chien N, Gibson KF, Raval JS, Saul M, Xue J, Zhang Y, Duncan SR. Autoantibody-targeted treatments for acute exacerbations of idiopathic pulmonary fibrosis. PLoS One. 2015;10(6):e0127771.CrossRef
Metadaten
Titel
Acute exacerbations of fibrosing interstitial lung disease associated with connective tissue diseases: a population-based study
verfasst von
Mengshu Cao
Jian Sheng
Xiaohua Qiu
Dandan Wang
Dongmei Wang
Yang Wang
Yonglong Xiao
Hourong Cai
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2019
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-019-0960-1

Weitere Artikel der Ausgabe 1/2019

BMC Pulmonary Medicine 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

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