Skip to main content
Erschienen in: Lung 5/2020

Open Access 25.08.2020 | IDIOPATHIC PULMONARY FIBROSIS

Blood Cell Count Derived Inflammation Indexes in Patients with Idiopathic Pulmonary Fibrosis

verfasst von: Angelo Zinellu, Panagiotis Paliogiannis, Elisabetta Sotgiu, Sabrina Mellino, Arduino A. Mangoni, Elisabetta Zinellu, Silvia Negri, Claudia Collu, Gianfranco Pintus, Antonello Serra, Angelo Maria Pistuddi, Ciriaco Carru, Pietro Pirina, Alessandro G. Fois

Erschienen in: Lung | Ausgabe 5/2020

Abstract

Purpose

Inflammation and immunity play a pivotal but yet unclear role in idiopathic pulmonary fibrosis (IPF), a chronic disorder characterized by progressive damage of lung parenchyma and severe loss of lung function despite optimal treatment. However, the pathophysiological and predictive role of combined blood cell count indexes of inflammation in IPF is uncertain.

Methods

Seventy-three patients with IPF and 62 healthy subjects matched for age, gender and smoking status were included in this cross-sectional study.

Results

We found significant differences in neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), monocyte to lymphocyte ratio (MLR), platelet to lymphocyte ratio (PLR), systemic inflammation response index (SIRI) and aggregate index of systemic inflammation (AISI) between IPF patients and healthy controls. In logistic regression, all combined blood inflammation indexes, barring PLR, were independently associated with the presence of IPF after adjusting for age, gender, body mass index and smoking status. Furthermore, significant associations between FVC% and NLR, LMR, SIRI and AISI, and between DLCO% and NLR, dNLR, LMR, SIRI and AISI, were observed.

Conclusions

In conclusion, our data indicate significant alterations of combined blood cell count indexes of inflammation in IPF.
Hinweise

Publisher's Note

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

Introduction

Idiopathic pulmonary fibrosis (IPF) is a chronic progressive fibrosing interstitial lung disease of unknown aetiology, occurring mainly in adults, that is characterized by a histopathologic and/or radiologic pattern of usual interstitial pneumonia [1]. The progressive accumulation of fibrotic tissue leads to irreversible lung damage. As a consequence, the estimated survival of untreated patients is limited, 2–3 years [2]. It is commonly accepted that fibrogenesis in IPF results from an aberrant repair process due to recurrent microinjury of alveolar epithelial cells [35]. The exaggerated wound repair and tissue remodelling lead to chronic inflammation and, ultimately, fibrosis. Both innate and adaptative responses seem to be involved in this process [6]. The excessive inflammatory response in IPF is multifactorial although the transforming growth factor-β (TGF-β) signalling is thought to play a key role [7]. It is well known that inflammation involves a highly coordinated network of immune-related cells, such as neutrophils, lymphocytes and monocytes [8]. Platelets can also modulate inflammation through direct cell–cell communications and the production of inflammatory mediators [9].
There is good evidence that indexes derived from calculating the ratios between neutrophils, lymphocytes, monocytes and platelets counts are more strongly associated with chronic inflammation conditions when compared with individual cell populations [8]. Indeed, combined blood cell count indexes of systemic inflammation have gained relevant scientific interest over the last decade. The neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), monocyte to lymphocyte ratio (MLR), platelet to lymphocyte ratio (PLR), systemic inflammation response index (SIRI) and aggregate inflammation systemic index (AISI) are increasingly being investigated as markers of inflammation in several disorders [1014], including lung disease states such as COPD [15, 16] and asthma [17]. However, no information is currently available regarding possible associations between combined blood cell count indexes of inflammation and IPF. We sought to address this issue by measuring NLR, dNLR, PLR, LMR SIRI and AISI in IPF patients and in controls and investigating associations with clinically relevant parameters of lung function, FEV1 (Forced Expiratory Volume in the 1st second), FVC (Forced Vital Capacity) and DLCO (Diffusion Capacity for Carbon Monoxide).

Materials and Methods

This was a case–control study of 73 consecutive newly diagnosed IPF patients recruited at the Respiratory Unit of the University of Sassari between 2016 and 2019. All patients signed a written consent and the study was approved by the ethics committee of the University Hospital of Cagliari (2262/CE-17/11/2015). IPF was diagnosed according to the current evidence-based guidelines for the diagnosis and management of IPF [18]. High-resolution computed tomography (HRCT) images and lung-biopsy samples of individual patients were reviewed by two experienced radiologists and two experienced pathologists, respectively. Each diagnosis was confirmed during multidisciplinary meetings attended by local respiratory, pathology, and radiology experts in interstitial lung disease. Measurements of FEV1, FVC and DLCO, expressed as percentages of predicted values (%FEV1, %FVC and %DLCO, respectively), were performed in accordance with the criteria published by the American Thoracic Society and the European Respiratory Society [19]. Patients with the following conditions were excluded from the study: patients currently experiencing an acute exacerbation of IPF; comorbid conditions including malignancy, bleeding tendency, and severe hepatic or renal dysfunction; use of immunosuppressants, antifibrotic drugs including interferon, d-penicillamine, and colchicine, or oral corticosteroids during the preceding 3 months. A group of 63 healthy controls matched for age, sex and smoking status, with no medical history, was also included.
Fasting blood samples from IPF patients and matched controls were respectively collected at first hospital admission in the Unit of Respiratory Diseases, and the Unit of Occupational Medicine of the University Hospital of Sassari (AOU Sassari); blood cell counts were performed using an automatic blood counter CellDyn Sapphire (Abbott Diagnostics, Santa Clara, CA, USA) in a certified laboratory. The following combined indexes were evaluated: NLR, dNLR, LMR, PLR, SIRI and AISI. The SIRI was calculated by multiplying the number of neutrophils with that of monocytes and dividing the product by the number of lymphocytes [20]. The AISI was calculated by multiplying the number of neutrophils, monocytes and platelets and dividing the product by the number of lymphocytes [10].
Results are expressed as mean values (mean ± SD) or median values (median and interquartile range, IQR). Variables distribution was assessed by the Shapiro–Wilk test. Statistical differences between groups were evaluated using unpaired Student’s t-test or Mann–Whitney rank sum test, as appropriate. Differences between categorical variables were assessed by Chi-squared test. Correlations between variables were assessed by Pearson’s correlation or Spearman’s correlation as appropriate. Logistic regression analysis was performed to evaluate independent associations between IPF disease and combined blood cell count indexes of inflammation. The association between different indexes and IPF was evaluated using receiver operating characteristics (ROC) curve analysis and selection of optimal cut-off values for sensitivity and specificity according to the Youden Index. Statistical analyses were performed using MedCalc for Windows, version 15.4 64 bit (MedCalc Software, Ostend, Belgium).

Results

Table 1 describes the clinical characteristics, lung functional parameters, and blood cell counts of IPF patients and healthy controls. As expected, FEV1 (%) and FVC (%) were significantly lower in IPF patients than in controls (mean 81 ± 22% vs 92 ± 18%, p < 0.001; median 72%; IQR 58–90% vs 92%; IQR 84–105% p < 0.001, respectively). IPF patients had higher WBCs (median 8.7 × 10L; IQR 7.3–10.3 × 10L vs 7.2 × 10L; IQR 6.4–8.7 × 10L, p < 0.001) and neutrophils (median 5.5 × 10L; IQR 4.6–6.7 × 10L vs 4.1 × 10L; IQR 3.5–5.4 × 10L, p < 0.001) and lower lymphocyte values (median 2.0 × 10L; IQ 1.5–2.4 × 10L vs 2.3 × 10L; IQR 1.9–2.6 × 10L, p < 0.05). There were no significant between-group differences in BMI, monocyte and platelet values.
Table 1
Demographic and functional parameters and blood count results of healthy subjects and IPF patients
 
Controls
(n = 62)
IPF
(n = 73)
Age (years)
67 (66–69)
70 (64–75)
Gender (F/M)
19/43
22/51
BMI (kg/m2)
27.1 ± 4.0
27.2 ± 3.9
Smokers, never/current/ex (%)
30/10/60
19/6/75
FEV1 (% predicted)
99 ± 18
81 ± 22***
FVC (% predicted)
92 (84–105)
72 (58–90)***
TLC (% predicted)
75 ± 17
DLCO (% predicted)
55 ± 23
6MWT (m)
342 ± 206
GAP index (1/2/3/4/5/6/7%)
4/19/25/24/17 /8/3
Disease stage (I/II/III %)
48/42/10
WBC (× 109 L)
7.2 (6.4–8.7)
8.7 (7.3–10.3)***
Monocytes (× 109 L)
0.5 (0.39–0.60)
0.5 (0.40–0.70)
Lymphocytes (× 109 L)
2.3 (1.9–2.6)
2.0 (1.5–2.4)*
Neutrophils, (× 109 L)
4.1 (3.5–5.4)
5.5 (4.6–6.7)***
Platelet (× 109 L)
242 ± 60
242 ± 68
Data are presented as mean ± standard deviation or median (interquartile range)
FEV1 forced expiratory volume in the 1st second, FVC forced vital capacity, TLC total lung capacity, DLCO diffusion capacity for carbon monoxide, GAP gender, age and two lung physiology variables index, P6MWT six-minute walk test, WBC white blood cell
*p < 0.05, **p < 0.01, ***p < 0.001
The combined indexes data are described in Table 2. NLR (median 2.67; IQR 1.89–2.67 vs 1.87; IQR 1.52–2.35, p < 0.001), d-NLR (median 1.88; IQR 1.33–2.26 vs 1.31; IQR 1.15–1.70, p < 0.001), PLR (mean 132 ± 74 vs 110 ± 36, p < 0.05), SIRI (median 1.49; IQR 0.89–2.40 vs 0.89; IQR 0.57–1.26, p < 0.001) and AISI (median 335; IQR 183–550 vs 198; IQR 128–316, p < 0.001) were significantly higher in IPF patients whereas the LMR was significantly lower (mean: 4.05± 1.75 vs 5.10 ± 1.71, p < 0.001). In univariate logistic regression, crude ORs for all inflammation combined indexes were significant, as reported in Table 3. In multivariate logistic regression, the ORs for NLR (OR = 2.633; 95% CI 1.604–4.322, p = 0.0001), d-NLR (OR = 3.941; 95% CI 1.851–8.390, p = 0.0004), LMR (OR = 0.685; 95%CI 0.543–0.865, p = 0.0015), SIRI (OR = 3.840; 95%CI 1.906–7.736, p = 0.0002) and AISI (OR = 1.003; 95% CI 1.001–1.005, p = 0.0045) remained significant after adjusting for age, gender, BMI and smoking status.
Table 2
Complete blood cell count-derived indexes in idiopathic pulmonary fibrosis and healthy subjects
 
Controls (62)
IPF (73)
NLR
1.87 (1.52–2.35)
2.67 (1.89–2.67)***
dNLR
1.31 (1.15–1.70)
1.88 (1.33–2.26)***
LMR
5.10 ± 1.71
4.05 ± 1.75c
PLR
110 ± 36
132 ± 74*
SIRI
0.89 (0.57–1.26)
1.49 (0.89–2.40)***
AISI
198 (128–316)
335 (183–550)***
Data are presented as mean ± standard deviation or median (interquartile range)
NLR neutrophil to lymphocyte, dNLR derived NLR, LMR lymphocyte to monocyte, PLR platelet to lymphocyte, SIRI Systemic Inflammation Response Index, AISI aggregate index of systemic inflammation, IPF idiopathic pulmonary fibrosis
*p < 0.05, **p < 0.01, ***p < 0.001
Table 3
Univariate and multivariate logistic regression for blood cell count–derived indexes
  
95% CI
p
Crude OR
 NLR
2.851
1.748–4.649
 < 0.0001
 dNLR
4.486
2.126–9.466
0.0001
 LMR
0.697
0.559–869
0.0013
 PLR
1.008
1.000–1.015
0.043
 SIRI
3.793
1.972–7.297
0.0001
 AISI
1.003
1.001–1.005
0.0014
Corrected OR
 NLR
2.633
1.604–4.322
0.0001
 dNLR
3.941
1.851–8.390
0.0004
 LMR
0.685
0.543–0.865
0.0015
 PLR
1.007
0.999–1.015
0.10
 SIRI
3.840
1.906–7.736
0.0002
 AISI
1.003
1.001–1.005
0.0045
NLR neutrophil to lymphocyte, dNLR derived NLR, LMR lymphocyte to monocyte, PLR platelet to lymphocyte, SIRI Systemic Inflammation Response Index, AISI aggregate index of systemic inflammation, OR odds ratio, CI confidence interval
In IPF patients, combined blood cell count indexes of inflammation were also significantly associated with functional lung parameters (Table 4). In particular, FEV1 was negatively associated with NLR (rho = − 0.24, p = 0.04), SIRI (rho = − 0.27, p = 0.019) and AISI (rho = − 0.26, p = 0.03), FVC% was negatively associated with NLR (rho = − 0.32, p = 0.007), SIRI (rho = − 0.30, p = 0.012) and AISI (rho = − 0.24 p = 0.04) and positively related with LMR (rho = 0.27, p = 0.02), and DLCO% was negatively associated with NLR (rho = − 0.40, p = 0.003), dNLR (rho = − 0.33, p = 0.015), SIRI (rho = − 0.35, p = 0.01) and AISI (rho = − 0.16, p = 0.26), and positively associated with LMR (rho = 0.28, p = 0.04). By contrast, barring LMR and SIRI, there were no significant associations between combined blood cell count indexes and functional lung parameters in healthy controls.
Table 4
Correlations between blood cell count indexes of systemic inflammation and functional parameters in patients with IPF and controls
 
Controls (n = 62)
IPF (n = 73)
FVC (%)
FEV1 (%)
FVC (%)
FEV1 (%)
DLCO (%)
NLR
rho = 0.04
p = 0.74
rho = −0.07
p = 0.60
rho = −0.32
p = 0.007
rho = −0.24
p = 0.04
rho = −0.40
p = 0.003
dNLR
rho = 0.08
p = 0.52
rho = 0.00
p = 0.96
rho = −0.18
p = 0.11
rho = −0.12
p = 0.31
rho = −0.33
p = 0.015
LMR
rho = 0.28
p = 0.03
rho = 0.39
p = 0.002
rho = 0.27
p = 0.02
rho = 0.22
p = 0.06
rho = 0.28
p = 0.04
PLR
rho = 0.21
p = 0.12
rho = 0.15
p = 0.24
rho = −0.19
p = 0.11
rho = −0.13
p = 0.27
rho = −0.16
p = 0.26
SIRI
rho = −0.24
p = 0.06
rho = −0.30
p = 0.017
rho = −0.30
p = 0.012
rho = −0.27
p = 0.019
rho = −0.35
p = 0.01
AISI
rho = −0.16
p = 0.21
rho = −0.25
p = 0.05
rho = −0.24
p = 0.04
rho = −0.26
p = 0.03
rho = −0.29
p = 0.04
NLR neutrophil to lymphocyte, dNLR derived NLR, LMR lymphocyte to monocyte, PLR platelet to lymphocyte, SIRI Systemic Inflammation Response Index, AISI Aggregate index of systemic inflammation, IPF idiopathic pulmonary fibrosis, FVC forced vital capacity, FEV1 forced expiratory volume in the 1st second, DLCO diffusion capacity for carbon monoxide
ROC curve analysis was carried out to evaluate the sensitivity, specificity, and the diagnostic accuracy of the combined inflammation indexes in distinguishing IPF patients from healthy subjects (Table 5). Except for PLR, all AUC values were significant. The better performing index was the NLR, with a threshold of 2.545, 55% sensitivity, and 85% specificity (AUC = 0.735, 95% CI 0.652–0.807 p < 0.001).
Table 5
Receiver operating characteristics (ROC) curves and prognostic accuracy of blood cell count–derived indexes
 
AUC
95% CI
p-value
Cut-off
Sensibility (%)
Specificity (%)
NLR
0.735
0.652–0.807
 < 0.0001
 > 2.545
55
85
dNLR
0.714
0.630–0.788
 < 0.0001
 > 1.862
53
85
LMR
0.663
0.576–0.742
0.0004
 < 2.857
29
100
PLR
0.590
0.502–0.674
0.07
 > 113
56
66
SIRI
0.729
0.646–0.802
 < 0.0001
 > 1.333
58
85
AISI
0.685
0.599–0.762
0.0001
 > 272
60
71
NLR neutrophil to lymphocyte, dNLR derived NLR, LMR lymphocyte to monocyte, PLR platelet to lymphocyte, SIRI Systemic Inflammation Response Index, AISI Aggregate index of systemic inflammation, AUC area under the curve, CI confidence interval

Discussion

We observed significant differences in neutrophil to lymphocyte ratio (NLR), derived neutrophil to lymphocyte ratio (dNLR), monocyte to lymphocyte ratio (MLR), platelet to lymphocyte ratio (PLR), systemic inflammation response index (SIRI) and aggregate index of systemic inflammation (AISI) between patients with IPF and a group of healthy controls matched for age, gender and smoking status. All combined blood inflammation indexes, except for PLR, were independently associated with IPF after adjusting for age, gender, body mass index and smoking status. Furthermore, there were significant associations between FVC% and NLR, LMR, SIRI and AISI, and between DLCO% and NLR, dNLR, LMR, SIRI and AISI.
IPF is the most severe and frequent form of interstitial lung disease, characterized by aberrant deposition of extracellular matrix due to recurrent micro-injuries to the alveolar epithelium. Mediators released by epithelial cells activate fibroblast proliferation leading to accumulation of fibroblasts and highly active myofibroblasts. These cells are resistant to apoptosis and lead to extensive lung remodelling, through deposition of extracellular matrix components such as hyaluronan, fibronectin and interstitial collagens, and consequent alveolar wall thickening and impaired gas exchange [6, 21]. While the exact aetiology of IPF remains unknown, several lines of evidence indicate that all stages of fibrosis are accompanied by innate and adaptive response, although treatment that modulates inflammation, e.g. steroids, has been shown to be ineffective, or even harmful, in clinical trials [2224]. Although this has led to the proposition that inflammation may represent an epiphenomenon, rather than a key driver in IPF, the active involvement of immune cells in the pathophysiology of the disease is well accepted.
Recent findings suggest that neutrophil elastase (NE) promotes TGF-β activation, fibroblast proliferation and myofibroblast differentiation, all contributing to enhance fibrosis [25]. The same study, according to previous observations [26], demonstrated that NE antagonists attenuate fibrosis both in asbestos and bleomycin induced fibrosis models, thus confirming the pro-fibrotic role of NE in IPF. Monocytes also play a crucial role during fibrogenesis as progenitor cells for pro-fibrotic macrophages and fibrocytes and by releasing pro-fibrotic inflammatory cytokines [7]. Furthermore, macrophages exhibit tissue-regenerating and pro-fibrotic function, depending on the local cytokine environment. In particular, M2 macrophages seem to have a central role in fibrosis regulation [27] acute exacerbations of IPF [28]. In addition, adaptive immunity cells Th1 and Th17 promote pulmonary fibrosis, whereas Th2 and Th22 cells inhibit fibrosis. Similarly, regulatory T cells (Tregs) and Th 9 cells exhibit both pro- and anti-fibrotic characteristics [29]. These observations strongly justify the investigation of blood cell count derived inflammation indexes in IPF.
Our results show significant alterations of these indexes in IPF patients, thus supporting the ability of these markers to detect systemic inflammation also in this group. These data agree with previous studies demonstrating that NLR and PLR are reliable indexes of systemic inflammation and predict the prognosis of several chronic inflammatory diseases [10, 11, 1517]. In logistic regression, except for PLR, all combined blood inflammation indexes were independently associated with the presence of IPF after adjusting for age, gender, BMI and smoking status. Moreover, they were significantly associated with robust functional lung parameters. In particular, the association between FVC% and NLR, LMR, SIRI and AISI and between DLCO% and NLR, dNLR, LMR, SIRI and AISI indicate a strong relationship between these inflammation indexes and disease severity. These data agree with previous observations in which the concentration of inflammatory biomarkers, such as BALF IL-8, was significantly associated with a reduced FVC% and DLCO% [30], and the extent of sputum neutrophilia was correlated with FVC% in IPF patients [31]. Furthermore, Tsoutsou et al. [32] found that serum concentrations of ICAM-2 (a circulating adhesion molecule with immunomodulatory effects) were negatively associated with DLCO%. Daniil et al. [33] also that reported that tissue CD8 + TLs inversely correlated with FVC%, TLC% and DLCO% and that CD68 + cells negative correlated with both FVC% and FEV1%.
In ROC curve analysis, the NLR, dNLR, LMR, SIRI and AISI were able to significantly discriminate patients with IPF from healthy subjects. The diagnostic accuracy was higher (AUC > 0.7) with NLR, dNLR and SIRI. NLR was the index which performed better, even than the combined indexes (SIRI and AISI) which included more than two blood cell populations in their calculation. The rationale for the use of such indexes, is based on the fact that multiple blood cell populations participate in systemic inflammation as we mentioned before, and therefore, inclusion of these populations in the calculation of an index should better reflect the inflammatory state and improve its predictive abilities. Indeed, the activation of neutrophils during systemic inflammation, not only increases their number, but stimulates also the production of several mediators (like IL-1a), which in turn stimulate megakaryocytes to produce platelets [34]; in addition, specific negligible fractions of monocytes in steady-state conditions have been reported to consistently increase during inflammation [35, 36]. Therefore, AISI or SIRI are hypothesized to perform better than more restricted indexes. Nevertheless, our study showed that both platelet and monocytes are not different between IPF patients and healthy controls, and combined indexes do not perform better than NLR. The latters have shown better predictive abilities than NLR, in acute clinical conditions and malignancies [10, 14, 37, 38]; this probably means that in cases of chronic inflammation adaptive phenomena prevent the increment of specific other than neutrophil and lymphocyte cell populations and stabilize their turnover. As a result, combined indexes may not be more effective than NLR in chronic systemic inflammation. The issue needs further evaluation. In addition, given the relatively low specificity of all the indexes tested in this study towards individual disease states, additional studies are required to determine their role in diagnosis, risk stratification, and prognosis, and their capacity to discriminate between IPF and other forms of interstitial lung diseases [1517].
Our study has some limitations. In particular, the relatively small sample size and the single centre nature of the study, even accounting for the low prevalence of the disease, limits its generalizability. Furthermore, the cross-sectional design did not allow establishing a definite cause-effect relationship between the studied indexes, the presence of IPF, and the lung function parameters and no correlations were analysed between these indexes and relevant clinical parameters, such as dyspnoea and the six-minute walking test. These issues notwithstanding, our data show the presence of significant alterations in NLR, dNLR, LMR, SIRI and AISI values in IPF patients when compared to healthy controls, and clinically relevant associations with lung function parameters. Such indexes might be attractive from a clinical point of view as their calculation is relatively simple, inexpensive, and part of routinely available laboratory investigations. Nevertheless, large, prospective studies are now required to investigate their prognostic role in IPF.

Compliance with Ethical Standards

Conflict of interest

The authors declare that they have no conflict of interest.

Ethical Approval

The study was approved by the ethics committee of the University Hospital of the University of Cagliari (2262/CE-17/11/2015).
Informed consent was obtained from all individual participants included in the study.
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/​.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Raghu G, Rochwerg B, Zhang Y et al (2015) An official ATS/ERS/JRS/ALAT clinical practice Guideline: treatment of idiopathic pulmonary fibrosis. An update of the 2011 clinical practice guideline. Am J Respir Crit Care Med 192:e3–19CrossRef Raghu G, Rochwerg B, Zhang Y et al (2015) An official ATS/ERS/JRS/ALAT clinical practice Guideline: treatment of idiopathic pulmonary fibrosis. An update of the 2011 clinical practice guideline. Am J Respir Crit Care Med 192:e3–19CrossRef
2.
Zurück zum Zitat du Bois RM (2012) An earlier and more confident diagnosis of idiopathic pulmonary fibrosis. Eur Respir Rev 21:141–146CrossRef du Bois RM (2012) An earlier and more confident diagnosis of idiopathic pulmonary fibrosis. Eur Respir Rev 21:141–146CrossRef
3.
Zurück zum Zitat Selman M, Pardo A (2002) Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder. Respir Res 3:3CrossRef Selman M, Pardo A (2002) Idiopathic pulmonary fibrosis: an epithelial/fibroblastic cross-talk disorder. Respir Res 3:3CrossRef
4.
Zurück zum Zitat Selman M, King TE, Pardo A et al (2001) (2001) Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 134:136–151CrossRef Selman M, King TE, Pardo A et al (2001) (2001) Idiopathic pulmonary fibrosis: prevailing and evolving hypotheses about its pathogenesis and implications for therapy. Ann Intern Med 134:136–151CrossRef
5.
Zurück zum Zitat Wuyts WA, Agostini C, Antoniou KM et al (2013) The pathogenesis of pulmonary fibrosis: a moving target. Eur Respir J 41:1207–1218CrossRef Wuyts WA, Agostini C, Antoniou KM et al (2013) The pathogenesis of pulmonary fibrosis: a moving target. Eur Respir J 41:1207–1218CrossRef
6.
Zurück zum Zitat Wynn TA (2011) Integrating mechanisms of pulmonary fibrosis. J Exp Med 208:1339–1350CrossRef Wynn TA (2011) Integrating mechanisms of pulmonary fibrosis. J Exp Med 208:1339–1350CrossRef
7.
Zurück zum Zitat Heukels P, Moor CC, von der Thüsen JH et al (2019) Inflammation and immunity in IPF pathogenesis and treatment. Respir Med 147:79–91CrossRef Heukels P, Moor CC, von der Thüsen JH et al (2019) Inflammation and immunity in IPF pathogenesis and treatment. Respir Med 147:79–91CrossRef
8.
Zurück zum Zitat Nathan C (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6:173–182CrossRef Nathan C (2006) Neutrophils and immunity: challenges and opportunities. Nat Rev Immunol 6:173–182CrossRef
9.
Zurück zum Zitat Karhausen J, Choi HW, Maddipati KR et al (2020) Platelets trigger perivascular mast cell degranulation to cause inflammatory responses and tissue injury. Sci Adv 6(12):eaay6314CrossRef Karhausen J, Choi HW, Maddipati KR et al (2020) Platelets trigger perivascular mast cell degranulation to cause inflammatory responses and tissue injury. Sci Adv 6(12):eaay6314CrossRef
10.
Zurück zum Zitat Paliogiannis P, Scognamillo F, Bellomo M et al (2015) Neutrophil to lymphocyte ratio as a predictor of thyroid papillary carcinoma. Acta Med Mediterr 31:371–375 Paliogiannis P, Scognamillo F, Bellomo M et al (2015) Neutrophil to lymphocyte ratio as a predictor of thyroid papillary carcinoma. Acta Med Mediterr 31:371–375
11.
Zurück zum Zitat Paliogiannis P, Satta R, Deligia G et al (2019) Associations between the neutrophil-to-lymphocyte and the platelet-to-lymphocyte ratios and the presence and severity of psoriasis: a systematic review and meta-analysis. Clin Exp Med 19:37–45CrossRef Paliogiannis P, Satta R, Deligia G et al (2019) Associations between the neutrophil-to-lymphocyte and the platelet-to-lymphocyte ratios and the presence and severity of psoriasis: a systematic review and meta-analysis. Clin Exp Med 19:37–45CrossRef
12.
Zurück zum Zitat Erre GL, Paliogiannis P, Castagna F et al (2019) Meta-analysis of neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio in rheumatoid arthritis. Eur J Clin Invest 49:e13037CrossRef Erre GL, Paliogiannis P, Castagna F et al (2019) Meta-analysis of neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio in rheumatoid arthritis. Eur J Clin Invest 49:e13037CrossRef
13.
Zurück zum Zitat Pinna A, Porcu T, D'Amico-Ricci G et al (2019) Complete blood cell count-derived inflammation biomarkers in men with age-related macular degeneration. Ocul Immunol Inflamm 27:932–936CrossRef Pinna A, Porcu T, D'Amico-Ricci G et al (2019) Complete blood cell count-derived inflammation biomarkers in men with age-related macular degeneration. Ocul Immunol Inflamm 27:932–936CrossRef
14.
Zurück zum Zitat Putzu C, Cortinovis DL, Colonese F et al (2018) Blood cell count indexes as predictors of outcomes in advanced non-small-cell lung cancer patients treated with nivolumab. Cancer Immunol Immunother 67:1349–1353CrossRef Putzu C, Cortinovis DL, Colonese F et al (2018) Blood cell count indexes as predictors of outcomes in advanced non-small-cell lung cancer patients treated with nivolumab. Cancer Immunol Immunother 67:1349–1353CrossRef
15.
Zurück zum Zitat Paliogiannis P, Fois AG, Sotgia S et al (2018) The neutrophil-to-lymphocyte ratio as a marker of chronic obstructive pulmonary disease and its exacerbations: a systematic review and meta-analysis. Eur J Clin Invest 48:e12984CrossRef Paliogiannis P, Fois AG, Sotgia S et al (2018) The neutrophil-to-lymphocyte ratio as a marker of chronic obstructive pulmonary disease and its exacerbations: a systematic review and meta-analysis. Eur J Clin Invest 48:e12984CrossRef
16.
Zurück zum Zitat Paliogiannis P, Fois AG, Sotgia S (2018) Neutrophil to lymphocyte ratio and clinical outcomes in COPD: recent evidence and future perspectives. Eur Respir Rev 27:147CrossRef Paliogiannis P, Fois AG, Sotgia S (2018) Neutrophil to lymphocyte ratio and clinical outcomes in COPD: recent evidence and future perspectives. Eur Respir Rev 27:147CrossRef
17.
Zurück zum Zitat Mochimaru T, Ueda S, Suzuki Y et al (2019) Neutrophil-to-lymphocyte ratio as a novel independent predictor of severe exacerbation in patients with asthma. Ann Allergy Asthma Immunol 122:337–339CrossRef Mochimaru T, Ueda S, Suzuki Y et al (2019) Neutrophil-to-lymphocyte ratio as a novel independent predictor of severe exacerbation in patients with asthma. Ann Allergy Asthma Immunol 122:337–339CrossRef
18.
Zurück zum Zitat Raghu G, Collard HR, Egan JJ et al (2011) An official ATS/ERS/JRS/ ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med 183:788–824CrossRef Raghu G, Collard HR, Egan JJ et al (2011) An official ATS/ERS/JRS/ ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med 183:788–824CrossRef
19.
Zurück zum Zitat Brusasco V, Crapo R, Viegi G et al (2005) Coming together: the ATS/ERS consensus on clinical pulmonary function testing. Eur Respir J 26:1–2CrossRef Brusasco V, Crapo R, Viegi G et al (2005) Coming together: the ATS/ERS consensus on clinical pulmonary function testing. Eur Respir J 26:1–2CrossRef
20.
Zurück zum Zitat Qi Q, Zhuang L, Shen Y et al (2016) A novel systemic inflammation response index (SIRI) for predicting the survival of patients with pancreatic cancer after chemotherapy. Cancer 122:2158–2167CrossRef Qi Q, Zhuang L, Shen Y et al (2016) A novel systemic inflammation response index (SIRI) for predicting the survival of patients with pancreatic cancer after chemotherapy. Cancer 122:2158–2167CrossRef
21.
Zurück zum Zitat Fernandez IE, Eickelberg O (2012) New cellular and molecular mechanisms of lung injury and fibrosis in idiopathic pulmonary fibrosis. Lancet 380:680–688CrossRef Fernandez IE, Eickelberg O (2012) New cellular and molecular mechanisms of lung injury and fibrosis in idiopathic pulmonary fibrosis. Lancet 380:680–688CrossRef
22.
Zurück zum Zitat King TE Jr, Albera C, Bradford WZ (2009) Effect of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis (INSPIRE): a multicentre, randomised, placebo-controlled trial. Lancet 374:222–228CrossRef King TE Jr, Albera C, Bradford WZ (2009) Effect of interferon gamma-1b on survival in patients with idiopathic pulmonary fibrosis (INSPIRE): a multicentre, randomised, placebo-controlled trial. Lancet 374:222–228CrossRef
23.
Zurück zum Zitat Raghu G, Brown KK, Bradford WZ et al (2004) A placebo-controlled trial of interferon gamma-1b in patients with idiopathic pulmonary fibrosis. N Engl J Med 350:125–133CrossRef Raghu G, Brown KK, Bradford WZ et al (2004) A placebo-controlled trial of interferon gamma-1b in patients with idiopathic pulmonary fibrosis. N Engl J Med 350:125–133CrossRef
24.
Zurück zum Zitat Raghu G, Brown KK, Collard HR et al (2017) Efficacy of simtuzumab versus placebo in patients with idiopathic pulmonary fibrosis: a randomised, double-blind, controlled, phase 2 trial. Lancet Respir Med 5:22–32CrossRef Raghu G, Brown KK, Collard HR et al (2017) Efficacy of simtuzumab versus placebo in patients with idiopathic pulmonary fibrosis: a randomised, double-blind, controlled, phase 2 trial. Lancet Respir Med 5:22–32CrossRef
25.
Zurück zum Zitat Gregory AD, Kliment CR, Metz HE et al (2015) Neutrophil elastase promotes myofibroblast differentiation in lung fibrosis. J Leukoc Biol 98:143–152CrossRef Gregory AD, Kliment CR, Metz HE et al (2015) Neutrophil elastase promotes myofibroblast differentiation in lung fibrosis. J Leukoc Biol 98:143–152CrossRef
26.
Zurück zum Zitat Takemasa A, Ishii Y, Fukuda T (2012) A neutrophil elastase inhibitor prevents bleomycin-induced pulmonary fibrosis in mice. Eur Respir J 40:1475–1482CrossRef Takemasa A, Ishii Y, Fukuda T (2012) A neutrophil elastase inhibitor prevents bleomycin-induced pulmonary fibrosis in mice. Eur Respir J 40:1475–1482CrossRef
27.
Zurück zum Zitat Mills CD (2012) M1 and M2 macrophages: oracles of health and disease. Crit Rev Immunol 32:463–488CrossRef Mills CD (2012) M1 and M2 macrophages: oracles of health and disease. Crit Rev Immunol 32:463–488CrossRef
28.
Zurück zum Zitat Schupp JC, Binder H, Jäger B et al (2015) Macrophage activation in acute exacerbation of idiopathic pulmonary fibrosis. PLoS ONE 10:e0116775CrossRef Schupp JC, Binder H, Jäger B et al (2015) Macrophage activation in acute exacerbation of idiopathic pulmonary fibrosis. PLoS ONE 10:e0116775CrossRef
29.
Zurück zum Zitat Kolahian S, Fernandez IE, Eickelberg O et al (2016) Immune mechanisms in pulmonary fibrosis. Am J Respir Cell Mol Biol 55:309–322CrossRef Kolahian S, Fernandez IE, Eickelberg O et al (2016) Immune mechanisms in pulmonary fibrosis. Am J Respir Cell Mol Biol 55:309–322CrossRef
30.
Zurück zum Zitat Ziegenhagen MW, Zabel P, Zissel G et al (1998) Serum level of interleukin 8 is elevated in idiopathic pulmonary fibrosis and indicates disease activity. Am J Respir Crit Care Med 157:762–768CrossRef Ziegenhagen MW, Zabel P, Zissel G et al (1998) Serum level of interleukin 8 is elevated in idiopathic pulmonary fibrosis and indicates disease activity. Am J Respir Crit Care Med 157:762–768CrossRef
31.
Zurück zum Zitat Beeh KM, Beier J, Kornmann O et al (2003) Neutrophilic inflammation in induced sputum of patients with idiopathic pulmonary fibrosis. Sarcoidosis Vasc Diffuse Lung Dis 20:138–143PubMed Beeh KM, Beier J, Kornmann O et al (2003) Neutrophilic inflammation in induced sputum of patients with idiopathic pulmonary fibrosis. Sarcoidosis Vasc Diffuse Lung Dis 20:138–143PubMed
32.
Zurück zum Zitat Tsoutsou PG, Gourgoulianis KI, Petinaki E et al (2004) ICAM-1, ICAM-2 and ICAM-3 in the sera of patients with idiopathic pulmonary fibrosis. Inflammation 28:359–364CrossRef Tsoutsou PG, Gourgoulianis KI, Petinaki E et al (2004) ICAM-1, ICAM-2 and ICAM-3 in the sera of patients with idiopathic pulmonary fibrosis. Inflammation 28:359–364CrossRef
33.
Zurück zum Zitat Daniil Z, Kitsanta P, Kapotsis G et al (2005) CD8+ T lymphocytes in lung tissue from patients with idiopathic pulmonary fibrosis. Respir Res 6:81CrossRef Daniil Z, Kitsanta P, Kapotsis G et al (2005) CD8+ T lymphocytes in lung tissue from patients with idiopathic pulmonary fibrosis. Respir Res 6:81CrossRef
34.
Zurück zum Zitat Nishimura S, Nagasaki M, Kunishima S et al (2015) IL-1alpha induces thrombopoiesis through megakaryocyte rupture in response to acute platelet needs. J Cell Biol 209:453–466CrossRef Nishimura S, Nagasaki M, Kunishima S et al (2015) IL-1alpha induces thrombopoiesis through megakaryocyte rupture in response to acute platelet needs. J Cell Biol 209:453–466CrossRef
35.
Zurück zum Zitat Yáñez A, Coetzee SG, Olsson A et al (2017) Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 47:890–902CrossRef Yáñez A, Coetzee SG, Olsson A et al (2017) Granulocyte-monocyte progenitors and monocyte-dendritic cell progenitors independently produce functionally distinct monocytes. Immunity 47:890–902CrossRef
36.
Zurück zum Zitat Satoh T, Nakagawa K, Sugihara F et al (2017) Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 541:96–101CrossRef Satoh T, Nakagawa K, Sugihara F et al (2017) Identification of an atypical monocyte and committed progenitor involved in fibrosis. Nature 541:96–101CrossRef
37.
Zurück zum Zitat Chen L, Kong X, Wang Z et al (2020) Pretreatment systemic inflammation response index in patients with breast cancer treated with neoadjuvant chemotherapy as a useful prognostic indicator. Cancer Manag Res 12:1543–1567CrossRef Chen L, Kong X, Wang Z et al (2020) Pretreatment systemic inflammation response index in patients with breast cancer treated with neoadjuvant chemotherapy as a useful prognostic indicator. Cancer Manag Res 12:1543–1567CrossRef
38.
Zurück zum Zitat Pacheco-Barcia V, Mondéjar Solís R, France T et al (2020) A systemic inflammation response index (SIRI) correlates with survival and predicts oncological outcome for mFOLFIRINOX therapy in metastatic pancreatic cancer. Pancreatology 20:254–264CrossRef Pacheco-Barcia V, Mondéjar Solís R, France T et al (2020) A systemic inflammation response index (SIRI) correlates with survival and predicts oncological outcome for mFOLFIRINOX therapy in metastatic pancreatic cancer. Pancreatology 20:254–264CrossRef
Metadaten
Titel
Blood Cell Count Derived Inflammation Indexes in Patients with Idiopathic Pulmonary Fibrosis
verfasst von
Angelo Zinellu
Panagiotis Paliogiannis
Elisabetta Sotgiu
Sabrina Mellino
Arduino A. Mangoni
Elisabetta Zinellu
Silvia Negri
Claudia Collu
Gianfranco Pintus
Antonello Serra
Angelo Maria Pistuddi
Ciriaco Carru
Pietro Pirina
Alessandro G. Fois
Publikationsdatum
25.08.2020
Verlag
Springer US
Erschienen in
Lung / Ausgabe 5/2020
Print ISSN: 0341-2040
Elektronische ISSN: 1432-1750
DOI
https://doi.org/10.1007/s00408-020-00386-7

Weitere Artikel der Ausgabe 5/2020

Lung 5/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

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

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Klimaschutz beginnt bei der Wahl des Inhalators

14.05.2024 Klimawandel Podcast

Auch kleine Entscheidungen im Alltag einer Praxis können einen großen Beitrag zum Klimaschutz leisten. Die neue Leitlinie zur "klimabewussten Verordnung von Inhalativa" geht mit gutem Beispiel voran, denn der Wechsel vom klimaschädlichen Dosieraerosol zum Pulverinhalator spart viele Tonnen CO2. Leitlinienautor PD Dr. Guido Schmiemann erklärt, warum nicht nur die Umwelt, sondern auch Patientinnen und Patienten davon profitieren.

Zeitschrift für Allgemeinmedizin, DEGAM

Typ-2-Diabetes und Depression folgen oft aufeinander

14.05.2024 Typ-2-Diabetes Nachrichten

Menschen mit Typ-2-Diabetes sind überdurchschnittlich gefährdet, in den nächsten Jahren auch noch eine Depression zu entwickeln – und umgekehrt. Besonders ausgeprägt ist die Wechselbeziehung laut GKV-Daten bei jüngeren Erwachsenen.

Update Innere Medizin

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