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

Open Access 01.12.2024 | Research

The effect of antifibrotic agents on acute respiratory failure in COVID-19 patients: a retrospective cohort study from TriNetX US collaborative networks

verfasst von: Hsin-Yi Wang, Shih-Chuan Tsai, Yi-Ching Lin, Jing-Uei Hou, Chih-Hao Chao

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2024

Abstract

Background

The coronavirus disease 2019 (COVID-19) pandemic has had a significant impact on global health and economies, resulting in millions of infections and deaths. This retrospective cohort study aimed to investigate the effect of antifibrotic agents (nintedanib and pirfenidone) on 1-year mortality in COVID-19 patients with acute respiratory failure.

Methods

Data from 61 healthcare organizations in the TriNetX database were analyzed. Adult patients with COVID-19 and acute respiratory failure were included. Patients with a pre-existing diagnosis of idiopathic pulmonary fibrosis before their COVID-19 diagnosis were excluded. The study population was divided into an antifibrotic group and a control group. Propensity score matching was used to compare outcomes, and hazard ratios (HR) for 1-year mortality were calculated.

Results

The antifibrotic group exhibited a significantly lower 1-year mortality rate compared to the control group. The survival probability at the end of the study was 84.42% in the antifibrotic group and 69.87% in the control group. The Log-Rank test yielded a p-value of less than 0.001. The hazard ratio was 0.434 (95% CI: 0.264–0.712), indicating a significant reduction in 1-year mortality in the antifibrotic group. Subgroup analysis demonstrated significantly improved 1-year survival in patients receiving nintedanib treatment and during periods when the Wuhan strain was predominant.

Discussion

This study is the first to demonstrate a survival benefit of antifibrotic agents in COVID-19 patients with acute respiratory failure. Further research and clinical trials are needed to confirm the efficacy of these antifibrotic agents in the context of COVID-19 and acute respiratory failure.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12890-024-02947-5.

Publisher’s Note

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

Background

Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus SARS-CoV-2, has had a major impact on global health and economies [1]. As of May 2023, there have been more than 760 million confirmed infections and 6.9 million deaths worldwide [2].
SARS-CoV-2 mainly infects the respiratory system and causes mild symptomatic or asymptomatic disease. However, approximately 14% of patients will require oxygen and hospital treatment, and approximately 5–6% will progress to severe pneumonia or acute respiratory distress syndrome (ARDS) and require intensive care unit (ICU) treatment [3]. The mortality rate in COVID-19 patients with ARDS requiring mechanical ventilation ranges from 35.7 to 94% [4, 5].
An emerging complication of COVID-19 is pulmonary fibrosis [6, 7]. Several studies have suggested that pulmonary fibrosis is a common complication that results in poor survival and functional outcomes in COVID-19 patients [8, 9]. Novel antifibrotic agents such as nintedanib and pirfenidone have demonstrated effectiveness in managing patients with idiopathic pulmonary fibrosis (IPF). Nintedanib functions as an intracellular inhibitor targeting vascular endothelial growth factor receptors 1–3, fibroblast growth factor receptors 1–3, and platelet-derived growth factor receptors a and b [10]. On the other hand, pirfenidone is an orally bioavailable pyridone derivative known for its anti-inflammatory, antifibrotic, and antioxidant properties [11]. Both medications have been proven to reduce the rate of annual decline in forced vital capacity and mortality in patients with IPF [1215]. Nintedanib also exhibits inhibitory effects on fibrogenesis across various pulmonary disorders, including connective tissue-associated interstitial lung diseases [16]. Notably, a recent study showed that antifibrotic therapy had similar efficacy in treating patients with progressive pulmonary fibrosis regardless of whether the underlying disease was IPF or non-IPF [17]. Novel antifibrotic agents are potential treatments for COVID-19-induced acute respiratory failure. We conducted this retrospective study to investigate the effect of antifibrotic agents on patients with acute respiratory failure due to COVID-19.

Methods

Setting

In this retrospective cohort study, we utilized the US Collaborative Network in the TriNetX database, which comprises 61 healthcare organizations (HCOs). TriNetX is a global federated health research network that provides access to electronic medical records, including diagnoses, procedures, medications, laboratory values, and genomic information across large HCOs.
The TriNetX platform complies with the Health Insurance Portability & Accountability Act and the General Data Protection Regulation. The Western Institutional Review Board has granted TriNetX a waiver of informed consent, as the platform only aggregates counts and statistical summaries of deidentified information.

Cohort

We included adult patients (≥ 20 years old) with a positive SARS-CoV-2 PCR test or a COVID-19 diagnosis (ICD10: U07.1, U07.2, or J12.82) during the study period in the TriNetX database. Patients with a past medical history of idiopathic pulmonary fibrosis (ICD10: J84.12) prior to their COVID-19 diagnosis were excluded. The index date was set as the day acute respiratory failure (ICD10: J96.00, J96.0, J96.01, and J96.02) developed within 3 days before to 1 month after the COVID-19 diagnosis. The study population was then divided into 2 groups: those who received oral antifibrotic treatment with nintedanib or pirfenidone (antifibrotic group) and those who did not receive antifibrotic treatment (control group). A flowchart of the cohort construction from participants enrolled between 1 June 2019 and 23 August 2023 is shown in Fig. 1. The main outcome of this study was 1-year mortality. Detailed query criteria are shown in Additional file 1.
Subgroup analysis was conducted to compare 1-year mortality rates between patients treated with nintedanib and those who received no antifibrotic agent, as well as between patients treated with pirfenidone and those who received no antifibrotic agent. Detailed query criteria are shown in Additional file 2 and 3. Additionally, we examined 1-year mortality rates between the antifibrotic and control groups using the same method but within different time frames to assess the effects of antifibrotic treatment across various COVID-19 strains. The study period was divided into four distinct time frames based on dominant strains in America: Whhan strain (December 2019 to April 2021, 88 patients), Alpha strain (May 2021 to June 2021, 29 patients), Delta strain (July 2021 to October 2021, 48 patients), and Omicron strain (November 2021 to August 2023, 95 patients). Detailed query criteria are shown in Additional file 4–7.

Statistical analyses

We performed propensity score matching at a 1:1 ratio on age at index, gender, comorbidities, and corticosteroid use. We used the TriNetX built-in function to compare outcomes. We calculated the hazard ratio (HR) of 1-year mortality for both groups. We tested the proportional hazard assumption using the generalized Schoenfeld approach built in the TriNetX platform. We used the Kaplan‒Meier method for the survival probability. We defined statistical significance as a p value < 0.05. A 95% confidence interval (95% CI) was also considered evidence of statistical significance.

Results

Baseline characteristics of the study population

Table 1 summarizes the demographic and lifestyle characteristics, comorbidities, adrenal corticosteroid, antiviral agents and biologic agents use in the antifibrotic and control groups before and after propensity score matching. The mean age of the population in both groups was approximately 64.5 years in the antifibrotic group and 65.8 years in the control group at the index date after matching. Approximately 59% of the individuals were male, and Caucasian was the predominant race (75.4% and 64.1% in the antifibrotic and control groups, respectively). The two groups were well matched regarding demographic, lifestyle, comorbidity characteristics. The propotion of patients receiving corticosteroid and remdesivir were similar in both groups. However, a significantly higher percentage of individuals in the antifibrotic group used Tocilizumab (6% vs. 0%, respectively, p = 0.001) (Table 1).
Table 1
Antifibrotic group (Cohort 1, N = 167) and Control group (cohort 2, N = 180,586) characteristics before and after propensity score matching
Before propensity score matching
After propensity score matching
 
Cohort
 
Patients
Mean ± SD
P-Value
Std diff.
Mean ± SD
Patients
P-Value
Std diff.
1
AI
Age at Index
167
64.5 +/- 11.6
0.126
0.137
64.5 +/- 11.6
167
100%
0.323
2
  
180,586
62.6 +/- 16.5
  
65.8 +/- 11.8
167
100%
 
Cohort
 
Patients
% of Cohort
P-Value
Std diff.
Patients
% of Cohort
P-Value
Std diff.
1
2106-3
White
126
75.4%
<0.001
0.288
126
75.4%
0.024
0.250
2
  
117,798
62.2%
  
107
64.1%
  
1
2054-5
Black or African
18
10.8%
0.132
0.124
18
10.8%
0.011
0.281
2
 
American
28,266
14.9%
  
35
21.0%
  
1
M
Male
99
59.3%
0.073
0.140
99
59.3%
1
<0.001
2
  
99,085
52.3%
  
99
59.3%
  
1
F
Female
64
38.3%
0.198
0.101
64
38.3%
1
<0.001
2
  
81,886
43.3%
  
64
38.3%
  
1
2028-9
Asian
10
6.0%
0.100
0.111
10
6.0%
1
<0.001
2
  
6,835
3.6%
  
10
6.0%
  
1
2131-1
Other Race
10
6.0%
0.129
0.104
10
6.0%
1
<0.001
2
  
7,106
3.8%
  
10
6.0%
  
Cohort
 
Patients
% of Cohort
P-Value
Std diff.
Patients
% of Cohort
P-Value
Std diff.
1
I20-I25
Ischemic heart
49
29.3%
0.003
0.215
49
29.3%
1
<0.001
2
 
diseases
38,072
20.1%
  
49
29.3%
  
1
E08-E13
Diabetes mellitus
46
27.5%
0.849
0.015
46
27.5%
1
<0.001
2
  
50,899
26.9%
  
46
27.5%
  
1
E40-E46
Malnutrition
11
6.6%
0.141
0.102
11
6.6%
0.822
0.025
2
  
8,104
4.3%
  
10
6.0%
  
1
C34
Malignant neoplasm of bronchus and lung
10
6.0%
<0.001
0.259
10
6.0%
1
<0.001
2
  
2,272
1.2%
  
10
6.0%
  
1
Z87.891
Personal history of nicotine dependence
39
23.4%
0.003
0.210
39
23.4%
0.282
0.118
2
  
28,627
15.1%
  
31
18.6%
  
1
Z72.0
Tobacco use
10
6.0%
0.059
0.125
10
6.0%
1
<0.001
2
  
6,347
3.4%
  
10
6.0%
  
1
K70
Alcoholic liver
10
6.0%
<0.001
0.286
10
6.0%
1
<0.001
2
 
disease
1,597
0.8%
  
10
6.0%
  
Cohort
 
Patients
% of Cohort
P-Value
Std diff.
Patients
% of Cohort
P-Value
Std diff.
1
HS050
ADRENAL
114
68.3%
<0.001
0.393
114
68.3%
1
<0.001
2
 
CORTICOSTEROIDS
93,286
49.3%
  
114
68.3%
  
1
3264
dexamethasone
61
36.5%
0.015
0.181
61
36.5%
0.219
0.135
2
  
53,188
28.1%
  
72
43.1%
  
1
6902
methylprednisolone
61
36.5%
<0.001
0.288
61
36.5%
0.246
0.127
2
  
44,376
23.4%
  
51
30.5%
  
1
5492
hydrocortisone
17
10.2%
0.680
0.031
17
10.2%
0.312
0.111
2
  
17,517
9.3%
  
23
13.8%
  
1
8640
prednisone
62
37.1%
<0.001
0.382
62
37.1%
0.164
0.153
2
  
38,179
20.2%
  
50
29.9%
  
1
2,284,718
remdesivir
10
6.0%
0.592
0.040
10
6.0%
1
<0.001
2
  
9,611
5.1%
  
10
6.0%
  
1
612,865
tocilizumab
10
6.0%
<0.001
0.322
10
6.0%
0.001
0.357
2
  
760
0.4%
  
0
0%
  
1
2,047,232
baricitinib
10
6.0%
<0.001
0.318
10
6.0%
1
<0.001
2
  
843
0.4%
  
10
6.0%
  

Mortality

Figure 2 shows the Kaplan‒Meier curve of survival probability. The survival probability at the end of the study was 84.42% in the antifibrotic group and 69.87% in the control group. The Log-Rank test yielded a p value of less than0.001. The hazard ratio was 0.434 (95% CI: 0.264–0.712). The chi-square was 6.721, and the p value was 0.01 for the proportionality assessment (Table 2). The raw data for the Kaplan-Meier graph was shown in additional file 8.
Table 2
Kaplan – Meier survival analysis of 1-year mortality
 
Patients in cohort
Patients with outcome
Median survival (days)
Survival probability at end of time window
Antifibrotic Group
166
25
--
84.42%
Control Group
163
42
--
69.87%
 
χ2
df
p
   
Log-Rank Test
11.556
1
< 0.001
   
 
Hazard Ratio
95% CI
χ2
df
p
Hazard Ratio and Proportionality
0.434
(0.264, 0.712)
6.721
1
0.010
Subgroup analysis revealed that out of the total participants, 127 patients received nintedanib, while 39 patients received pirfenidone. We observed a statistically significant improvement in 1-year mortality rates in the nintedanib group compared to the control group (p = 0.013). Although there was a noted improvement in the pirfenidone group, it was not statistically significant (p = 0.601) (Fig. 3a and b). The raw data for the Kaplan-Meier graph was shown in additional file 9 and 10.
After dividing the study period into four distinct time frames, our analysis indicated better survival rates in the antifibrotic group across all time frames, with statistical significance observed only in the Wuhan strain period (p = 0.002) (Fig. 4a-d). The raw data for the Kaplan-Meier graph was shown in additional file 11–14.

Discussion

The present retrospective multi-institutional study was conducted by the U.S. The Collaborative Research Network has demonstrated that the novel antifibrotic agents nintedanib and pirfenidone effectively reduce the 1-year mortality rate among COVID-19 patients suffering from acute respiratory failure. Remarkably, this study represents, to our knowledge, the first instance of survival benefit observed.
We observed significantly better 1-year survival in patient receiving nintedanib compared to those receiving pirfenidone. However, only 39 patients received pirfenidone in our cohort, indicating that small sample size may have contributed to insignificant results. Likewise, the small sample size during Alpha strain and Delta strain periods could also have led to insignificant findings. The characteristics of the Omicron strain, including its tendency to infect the upper respiratory tract rather than the lower respiratory tract and its lower IL-6 secretion, may have influenced the effectiveness of antifibrotic agents [18]. However, due to limitations of the TriNetX database, which does not specify the virus strain patients were infected with, comparing the effects of antifibrotic treatment across different strains was challenging. There likely were cases of mixed-strain infection across the four time frames. As a result, we are unable to ascertain the actual effect of antifibrotic agents on different virus strain. Further studies are warranted to validate these findings.
Our result revealed a higher percentage of patients in the antifibrotic group received Tocilizumab. Elevated IL-6 concentrations are associated with severe outcomes in COVID-19 [19]. Tocilizumab, an IL-6 receptor antagonist, has been demonstrated to reduce inflammatory responses and improve 28-day mortality in COVID-19 patients requiring oxygen therapy [20].Currently, the World Health Organization suggested using Tocilizumab in severe or critical COVID-19 patients who exhibit signs of desaturation in room air, severe respiratory distress, ARDS, require life-sustaining treatment, sepsis and septic shock [21]. However, Tocilizumab may also exacerbate bacterial infections in COVID-19 patients, which could restrict its usage. In the RECOVERY trial, 16% of patients in the Tocilizumab group ultimately did not receive this treatment [20]. Tocilizumab would be administered only to COVID-19 patients with higher severity and fewer signs of complicating bacterial infection. This observation may reflect a higher severity of illness among patients in the antifibrotic group.
Nintedanib functions by binding to intracellular ATP pockets and inhibiting profibrotic signaling pathways, including platelet-derived growth factor, fibroblast growth factor, vascular endothelial growth factor, and transforming growth factor-beta (TGF-β) [10]. Similarly, pirfenidone regulates pro-fibrotic and pro-inflammatory cytokines such as TGF-β, tumor necrosis factor-α, interferon γ, interleukin-1β, and interleukin-6, thus inhibiting fibroblast proliferation and collagen synthesis [22, 23]. Moreover, pirfenidone has demonstrated a capacity to reduce ACE receptor expression, which is considered a major cellular receptor for SARS-CoV-2 virus entry [24]. Notably, case reports have indicated successful treatment outcomes using nintedanib for COVID-19-related ARDS [25, 26]. While ongoing studies on the treatment of COVID-19 patients with acute respiratory failure or pulmonary fibrosis exist, only limited published data are currently available. Umemura et al. observed that nintedanib significantly shortened the duration of mechanical ventilation and reduced the high attenuation area percentage on computed tomography volumetry in COVID-19 patients admitted to the ICU and requiring mechanical ventilation. However, no significant differences in 28-day mortality were found [27]. Similarly, Zhang et al. demonstrated that pirfenidone significantly decreased inflammatory biomarkers, including interleukin-2R, tumor necrosis factor-alpha (TNF-α), and D-dimer, although clinical parameters such as clinical improvement time, duration of oxygen therapy, time from randomization to death, and interstitial changes on CT images exhibited insignificant improvement [28].
Epithelial injury followed by a subsequent fibroproliferative cascade has been recognized as a key pathogenic mechanism of pulmonary fibrosis shared between COVID-19-related ARDS and idiopathic pulmonary fibrosis [2932]. Type 2 alveolar epithelial cells (ATII cells) plays a crucial role in IPF development [33, 34, 35, 36]. SARS-CoV-2 enters lower respiratory tract cells via the angiotensin-converting enzyme 2 (ACE2) receptor in conjunction with transmembrane protease serine 2, expressed by ATII cells. Inadequate responses of ATII cells to lung injury lead to aberrant tissue repair, characterized by fibroblast activation, collagen deposition, connective tissue accumulation, and angiogenesis [37, 38]. Both idiopathic pulmonary fibrosis and COVID-19-induced pulmonary fibrosis involve the renin-angiotensin system (RAS) in their disease progression. A pivotal player in this context, ACE2, interacts with other components of the RAS. ACE2-mediated SARS-CoV-2 entry into lung cells is believed to result in reduced ACE2 expression, disturbing the RAS system balance and subsequently triggering inflammation and fibrosis [39].
Dysregulation of microRNAs (miRNAs) has been implicated in the development of pulmonary fibrosis in COVID-19 patients, contributing to collagen deposition and myofibroblast transformation [40]. This miRNA imbalance has also been observed in idiopathic pulmonary fibrosis patients [41]. Lacedonia et al. analyzed the expression of exosomal miRNAs and confirmed the key involvement of a let-7d down-regulation and dysregulation of miR-16 in IPF [42]. Notably, Guiot et al. identified a total of 34 dysregulated miRNAs that overlapped between COVID-19 and idiopathic pulmonary fibrosis [40].
Despite the insightful findings, our study has certain limitations. The utilization of retrospective electronic records introduces inherent weaknesses. Lack of access to raw data prevents an accurate assessment of disease severity, identification of the causes of acute respiratory failure, determination of whether SARS-CoV-2 related pneumonia was presenting, specification of specific dosage and timing or the reasons for initiating antifibrotic treatment. Potential miscoding, inaccurate coding, or incomplete clinical information about comorbidities, socioeconomic status, and lifestyle habits may also introduce biases. Moreover, the TriNetX database lacks certain clinical information, such as ventilator-free days, ICU days, ventilator weaning rates, and hospitalization duration. Consequently, the efficacy of antifibrotic therapy cannot be adequately evaluated based on these parameters.

Conclusions

This retrospective multi-institutional study highlights the potential benefits of novel antifibrotic agents, nintedanib and pirfenidone, in reducing mortality among COVID-19 patients with acute respiratory failure. This study offers important insights into the therapeutic potential of these agents in managing the complex pathogenesis of both COVID-19-induced pulmonary fibrosis and idiopathic pulmonary fibrosis. The findings underscore the significance of targeting fibroproliferative pathways and the RAS in mitigating inflammation and fibrosis. However, it is important to acknowledge the study’s limitations, particularly the retrospective nature of data analysis and the potential for biases. Future research and clinical trials are needed to further validate the efficacy of these antifibrotic agents and explore their precise mechanisms of action in the context of COVID-19 and acute respiratory failure.

Acknowledgements

Not applicable.

Declarations

The use of TriNetX for this study was approved under the authority of the Institutional Review Board of Taichung Veterans General Hospital (TCVGH-IRB No. SE22220A-1). The Western Institutional Review Board has granted TriNetX a waiver of informed consent, as the platform only aggregates counts and statistical summaries of deidentified information.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Lai CC, Shih TP, Ko WC, Tang HJ, Hsueh PR. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): the epidemic and the challenges. Int J Antimicrob Agents. 2020;55(3):105924.CrossRefPubMedPubMedCentral Lai CC, Shih TP, Ko WC, Tang HJ, Hsueh PR. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease-2019 (COVID-19): the epidemic and the challenges. Int J Antimicrob Agents. 2020;55(3):105924.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of Coronavirus Disease 2019 in China. N Engl J Med. 2020;382(18):1708–20.CrossRefPubMed Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al. Clinical characteristics of Coronavirus Disease 2019 in China. N Engl J Med. 2020;382(18):1708–20.CrossRefPubMed
4.
Zurück zum Zitat Auld SC, Caridi-Scheible M, Blum JM, Robichaux C, Kraft C, Jacob JT, et al. ICU and ventilator mortality among critically ill adults with Coronavirus Disease 2019. Crit Care Med. 2020;48(9):e799–804.CrossRefPubMed Auld SC, Caridi-Scheible M, Blum JM, Robichaux C, Kraft C, Jacob JT, et al. ICU and ventilator mortality among critically ill adults with Coronavirus Disease 2019. Crit Care Med. 2020;48(9):e799–804.CrossRefPubMed
5.
Zurück zum Zitat Sinha S, Sardesai I, Galwankar SC, Nanayakkara PWB, Narasimhan DR, Grover J, et al. Optimizing respiratory care in coronavirus disease-2019: a comprehensive, protocolized, evidence-based, algorithmic approach. Int J Crit Illn Inj Sci. 2020;10(2):56–63.CrossRefPubMedPubMedCentral Sinha S, Sardesai I, Galwankar SC, Nanayakkara PWB, Narasimhan DR, Grover J, et al. Optimizing respiratory care in coronavirus disease-2019: a comprehensive, protocolized, evidence-based, algorithmic approach. Int J Crit Illn Inj Sci. 2020;10(2):56–63.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and COVID-19: the potential role for antifibrotic therapy. Lancet Respir Med. 2020;8(8):807–15.CrossRefPubMedPubMedCentral George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and COVID-19: the potential role for antifibrotic therapy. Lancet Respir Med. 2020;8(8):807–15.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat John AE, Joseph C, Jenkins G, Tatler AL. COVID-19 and pulmonary fibrosis: a potential role for lung epithelial cells and fibroblasts. Immunol Rev. 2021;302(1):228–40.CrossRefPubMedPubMedCentral John AE, Joseph C, Jenkins G, Tatler AL. COVID-19 and pulmonary fibrosis: a potential role for lung epithelial cells and fibroblasts. Immunol Rev. 2021;302(1):228–40.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Pan Y, Guan H, Zhou S, Wang Y, Li Q, Zhu T, et al. Initial CT findings and temporal changes in patients with the novel coronavirus pneumonia (2019-nCoV): a study of 63 patients in Wuhan, China. Eur Radiol. 2020;30(6):3306–9.CrossRefPubMedPubMedCentral Pan Y, Guan H, Zhou S, Wang Y, Li Q, Zhu T, et al. Initial CT findings and temporal changes in patients with the novel coronavirus pneumonia (2019-nCoV): a study of 63 patients in Wuhan, China. Eur Radiol. 2020;30(6):3306–9.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Schwensen HF, Borreschmidt LK, Storgaard M, Redsted S, Christensen S, Madsen LB. Fatal pulmonary fibrosis: a post-COVID-19 autopsy case. J Clin Pathol. 2020. Schwensen HF, Borreschmidt LK, Storgaard M, Redsted S, Christensen S, Madsen LB. Fatal pulmonary fibrosis: a post-COVID-19 autopsy case. J Clin Pathol. 2020.
10.
Zurück zum Zitat Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45(5):1434–45.CrossRefPubMedPubMedCentral Wollin L, Wex E, Pautsch A, Schnapp G, Hostettler KE, Stowasser S, et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45(5):1434–45.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Conte E, Gili E, Fagone E, Fruciano M, Iemmolo M, Vancheri C. Effect of pirfenidone on proliferation, TGF-beta-induced myofibroblast differentiation and fibrogenic activity of primary human lung fibroblasts. Eur J Pharm Sci. 2014;58:13–9.CrossRefPubMed Conte E, Gili E, Fagone E, Fruciano M, Iemmolo M, Vancheri C. Effect of pirfenidone on proliferation, TGF-beta-induced myofibroblast differentiation and fibrogenic activity of primary human lung fibroblasts. Eur J Pharm Sci. 2014;58:13–9.CrossRefPubMed
12.
Zurück zum Zitat Richeldi L, Costabel U, Selman M, Kim DS, Hansell DM, Nicholson AG, et al. Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. N Engl J Med. 2011;365(12):1079–87.CrossRefPubMed Richeldi L, Costabel U, Selman M, Kim DS, Hansell DM, Nicholson AG, et al. Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. N Engl J Med. 2011;365(12):1079–87.CrossRefPubMed
13.
Zurück zum Zitat Richeldi L, Kreuter M, Selman M, Crestani B, Kirsten AM, Wuyts WA, et al. Long-term treatment of patients with idiopathic pulmonary fibrosis with nintedanib: results from the TOMORROW trial and its open-label extension. Thorax. 2018;73(6):581–3.CrossRefPubMed Richeldi L, Kreuter M, Selman M, Crestani B, Kirsten AM, Wuyts WA, et al. Long-term treatment of patients with idiopathic pulmonary fibrosis with nintedanib: results from the TOMORROW trial and its open-label extension. Thorax. 2018;73(6):581–3.CrossRefPubMed
14.
Zurück zum Zitat King TE Jr., Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370(22):2083–92.CrossRefPubMed King TE Jr., Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, et al. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370(22):2083–92.CrossRefPubMed
15.
Zurück zum Zitat Taguchi Y, Ebina M, Hashimoto S, Ogura T, Azuma A, Taniguchi H, et al. Efficacy of pirfenidone and disease severity of idiopathic pulmonary fibrosis: extended analysis of phase III trial in Japan. Respir Investig. 2015;53(6):279–87.CrossRefPubMed Taguchi Y, Ebina M, Hashimoto S, Ogura T, Azuma A, Taniguchi H, et al. Efficacy of pirfenidone and disease severity of idiopathic pulmonary fibrosis: extended analysis of phase III trial in Japan. Respir Investig. 2015;53(6):279–87.CrossRefPubMed
16.
Zurück zum Zitat Flaherty KR, Wells AU, Cottin V, Devaraj A, Walsh SLF, Inoue Y, et al. Nintedanib in Progressive Fibrosing interstitial lung diseases. N Engl J Med. 2019;381(18):1718–27.CrossRefPubMed Flaherty KR, Wells AU, Cottin V, Devaraj A, Walsh SLF, Inoue Y, et al. Nintedanib in Progressive Fibrosing interstitial lung diseases. N Engl J Med. 2019;381(18):1718–27.CrossRefPubMed
17.
Zurück zum Zitat Finnerty JP, Ponnuswamy A, Dutta P, Abdelaziz A, Kamil H. Efficacy of antifibrotic drugs, nintedanib and pirfenidone, in treatment of progressive pulmonary fibrosis in both idiopathic pulmonary fibrosis (IPF) and non-IPF: a systematic review and meta-analysis. BMC Pulm Med. 2021;21(1):411.CrossRefPubMedPubMedCentral Finnerty JP, Ponnuswamy A, Dutta P, Abdelaziz A, Kamil H. Efficacy of antifibrotic drugs, nintedanib and pirfenidone, in treatment of progressive pulmonary fibrosis in both idiopathic pulmonary fibrosis (IPF) and non-IPF: a systematic review and meta-analysis. BMC Pulm Med. 2021;21(1):411.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zaderer V, Abd El Halim H, Wyremblewsky AL, Lupoli G, Dachert C, Muenchhoff M, et al. Omicron subvariants illustrate reduced respiratory tissue penetration, cell damage and inflammatory responses in human airway epithelia. Front Immunol. 2023;14:1258268.CrossRefPubMedPubMedCentral Zaderer V, Abd El Halim H, Wyremblewsky AL, Lupoli G, Dachert C, Muenchhoff M, et al. Omicron subvariants illustrate reduced respiratory tissue penetration, cell damage and inflammatory responses in human airway epithelia. Front Immunol. 2023;14:1258268.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Del Valle DM, Kim-Schulze S, Huang HH, Beckmann ND, Nirenberg S, Wang B, et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med. 2020;26(10):1636–43.CrossRefPubMedPubMedCentral Del Valle DM, Kim-Schulze S, Huang HH, Beckmann ND, Nirenberg S, Wang B, et al. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med. 2020;26(10):1636–43.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Group RC. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397(10285):1637–45.CrossRef Group RC. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet. 2021;397(10285):1637–45.CrossRef
21.
Zurück zum Zitat Agarwal A, Hunt BJ, Stegemann M, Rochwerg B, Lamontagne F, Siemieniuk RA, et al. A living WHO guideline on drugs for covid-19. BMJ. 2020;370:m3379.CrossRefPubMed Agarwal A, Hunt BJ, Stegemann M, Rochwerg B, Lamontagne F, Siemieniuk RA, et al. A living WHO guideline on drugs for covid-19. BMJ. 2020;370:m3379.CrossRefPubMed
22.
Zurück zum Zitat Raghu G, Richeldi L. Current approaches to the management of idiopathic pulmonary fibrosis. Respir Med. 2017;129:24–30.CrossRefPubMed Raghu G, Richeldi L. Current approaches to the management of idiopathic pulmonary fibrosis. Respir Med. 2017;129:24–30.CrossRefPubMed
23.
Zurück zum Zitat Yue X, Shan B, Lasky JA. TGF-beta: Titan of Lung Fibrogenesis. Curr Enzym Inhib. 2010;6(2). Yue X, Shan B, Lasky JA. TGF-beta: Titan of Lung Fibrogenesis. Curr Enzym Inhib. 2010;6(2).
24.
Zurück zum Zitat Li C, Han R, Kang L, Wang J, Gao Y, Li Y, et al. Pirfenidone controls the feedback loop of the AT1R/p38 MAPK/renin-angiotensin system axis by regulating liver X receptor-alpha in myocardial infarction-induced cardiac fibrosis. Sci Rep. 2017;7:40523.CrossRefPubMedPubMedCentral Li C, Han R, Kang L, Wang J, Gao Y, Li Y, et al. Pirfenidone controls the feedback loop of the AT1R/p38 MAPK/renin-angiotensin system axis by regulating liver X receptor-alpha in myocardial infarction-induced cardiac fibrosis. Sci Rep. 2017;7:40523.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Marwah V, Choudhary R, Bhati G, Peter DK. Early experience with anti-interleukin-6 therapy in COVID-19 hyperinflammation. Lung India. 2021;38(Supplement):S119–21.PubMedPubMedCentral Marwah V, Choudhary R, Bhati G, Peter DK. Early experience with anti-interleukin-6 therapy in COVID-19 hyperinflammation. Lung India. 2021;38(Supplement):S119–21.PubMedPubMedCentral
26.
Zurück zum Zitat Ogata H, Nakagawa T, Sakoda S, Ishimatsu A, Taguchi K, Kadowaki M, et al. Nintedanib treatment for pulmonary fibrosis after coronavirus disease 2019. Respirol Case Rep. 2021;9(5):e00744.CrossRefPubMedPubMedCentral Ogata H, Nakagawa T, Sakoda S, Ishimatsu A, Taguchi K, Kadowaki M, et al. Nintedanib treatment for pulmonary fibrosis after coronavirus disease 2019. Respirol Case Rep. 2021;9(5):e00744.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Umemura Y, Mitsuyama Y, Minami K, Nishida T, Watanabe A, Okada N, et al. Efficacy and safety of nintedanib for pulmonary fibrosis in severe pneumonia induced by COVID-19: an interventional study. Int J Infect Dis. 2021;108:454–60.CrossRefPubMedPubMedCentral Umemura Y, Mitsuyama Y, Minami K, Nishida T, Watanabe A, Okada N, et al. Efficacy and safety of nintedanib for pulmonary fibrosis in severe pneumonia induced by COVID-19: an interventional study. Int J Infect Dis. 2021;108:454–60.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Zhang FWY, He L, Zhang H, Hu Q, Yue H, He J, Dai H. A trial of pirfenidone in hospitalized adult patients with severe coronavirus disease 2019. Chin Med J (Engl). 2022;135:368–70.CrossRef Zhang FWY, He L, Zhang H, Hu Q, Yue H, He J, Dai H. A trial of pirfenidone in hospitalized adult patients with severe coronavirus disease 2019. Chin Med J (Engl). 2022;135:368–70.CrossRef
29.
Zurück zum Zitat Wijsenbeek M, Cottin V. Spectrum of Fibrotic Lung diseases. N Engl J Med. 2020;383(10):958–68.CrossRefPubMed Wijsenbeek M, Cottin V. Spectrum of Fibrotic Lung diseases. N Engl J Med. 2020;383(10):958–68.CrossRefPubMed
30.
Zurück zum Zitat Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389(10082):1941–52.CrossRefPubMed Richeldi L, Collard HR, Jones MG. Idiopathic pulmonary fibrosis. Lancet. 2017;389(10082):1941–52.CrossRefPubMed
31.
Zurück zum Zitat Lee JM, Yoshida M, Kim MS, Lee JH, Baek AR, Jang AS, et al. Involvement of alveolar epithelial cell necroptosis in idiopathic pulmonary fibrosis pathogenesis. Am J Respir Cell Mol Biol. 2018;59(2):215–24.CrossRefPubMed Lee JM, Yoshida M, Kim MS, Lee JH, Baek AR, Jang AS, et al. Involvement of alveolar epithelial cell necroptosis in idiopathic pulmonary fibrosis pathogenesis. Am J Respir Cell Mol Biol. 2018;59(2):215–24.CrossRefPubMed
32.
Zurück zum Zitat Minagawa S, Yoshida M, Araya J, Hara H, Imai H, Kuwano K. Regulated necrosis in Pulmonary Disease. A focus on necroptosis and Ferroptosis. Am J Respir Cell Mol Biol. 2020;62(5):554–62.CrossRefPubMed Minagawa S, Yoshida M, Araya J, Hara H, Imai H, Kuwano K. Regulated necrosis in Pulmonary Disease. A focus on necroptosis and Ferroptosis. Am J Respir Cell Mol Biol. 2020;62(5):554–62.CrossRefPubMed
33.
Zurück zum Zitat Confalonieri P, Volpe MC, Jacob J, Maiocchi S, Salton F, Ruaro B et al. Regeneration or repair? The role of alveolar epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cells. 2022;11(13). Confalonieri P, Volpe MC, Jacob J, Maiocchi S, Salton F, Ruaro B et al. Regeneration or repair? The role of alveolar epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cells. 2022;11(13).
34.
35.
Zurück zum Zitat Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, et al. SARS-CoV-2 receptor ACE2 is an Interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020;181(5):1016–e3519.CrossRefPubMedPubMedCentral Ziegler CGK, Allon SJ, Nyquist SK, Mbano IM, Miao VN, Tzouanas CN, et al. SARS-CoV-2 receptor ACE2 is an Interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell. 2020;181(5):1016–e3519.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Carcaterra M, Caruso C. Alveolar epithelial cell type II as main target of SARS-CoV-2 virus and COVID-19 development via NF-Kb pathway deregulation: a physio-pathological theory. Med Hypotheses. 2021;146:110412.CrossRefPubMed Carcaterra M, Caruso C. Alveolar epithelial cell type II as main target of SARS-CoV-2 virus and COVID-19 development via NF-Kb pathway deregulation: a physio-pathological theory. Med Hypotheses. 2021;146:110412.CrossRefPubMed
37.
Zurück zum Zitat Parimon T, Yao C, Stripp BR, Noble PW, Chen P. Alveolar epithelial type II cells as drivers of lung fibrosis in idiopathic pulmonary fibrosis. Int J Mol Sci. 2020;21(7). Parimon T, Yao C, Stripp BR, Noble PW, Chen P. Alveolar epithelial type II cells as drivers of lung fibrosis in idiopathic pulmonary fibrosis. Int J Mol Sci. 2020;21(7).
38.
Zurück zum Zitat Selman M, Pardo A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal. 2020;66:109482.CrossRefPubMed Selman M, Pardo A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal. 2020;66:109482.CrossRefPubMed
39.
Zurück zum Zitat Tan WSD, Liao W, Zhou S, Mei D, Wong WF. Targeting the renin-angiotensin system as novel therapeutic strategy for pulmonary diseases. Curr Opin Pharmacol. 2018;40:9–17.CrossRefPubMed Tan WSD, Liao W, Zhou S, Mei D, Wong WF. Targeting the renin-angiotensin system as novel therapeutic strategy for pulmonary diseases. Curr Opin Pharmacol. 2018;40:9–17.CrossRefPubMed
40.
Zurück zum Zitat Guiot J, Henket M, Remacle C, Cambier M, Struman I, Winandy M, et al. Systematic review of overlapping microRNA patterns in COVID-19 and idiopathic pulmonary fibrosis. Respir Res. 2023;24(1):112.CrossRefPubMedPubMedCentral Guiot J, Henket M, Remacle C, Cambier M, Struman I, Winandy M, et al. Systematic review of overlapping microRNA patterns in COVID-19 and idiopathic pulmonary fibrosis. Respir Res. 2023;24(1):112.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Bagnato G, Roberts WN, Roman J, Gangemi S. A systematic review of overlapping microRNA patterns in systemic sclerosis and idiopathic pulmonary fibrosis. Eur Respir Rev. 2017;26(144). Bagnato G, Roberts WN, Roman J, Gangemi S. A systematic review of overlapping microRNA patterns in systemic sclerosis and idiopathic pulmonary fibrosis. Eur Respir Rev. 2017;26(144).
42.
Zurück zum Zitat Lacedonia D, Scioscia G, Soccio P, Conese M, Catucci L, Palladino GP, et al. Downregulation of exosomal let-7d and miR-16 in idiopathic pulmonary fibrosis. BMC Pulm Med. 2021;21(1):188.CrossRefPubMedPubMedCentral Lacedonia D, Scioscia G, Soccio P, Conese M, Catucci L, Palladino GP, et al. Downregulation of exosomal let-7d and miR-16 in idiopathic pulmonary fibrosis. BMC Pulm Med. 2021;21(1):188.CrossRefPubMedPubMedCentral
Metadaten
Titel
The effect of antifibrotic agents on acute respiratory failure in COVID-19 patients: a retrospective cohort study from TriNetX US collaborative networks
verfasst von
Hsin-Yi Wang
Shih-Chuan Tsai
Yi-Ching Lin
Jing-Uei Hou
Chih-Hao Chao
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2024
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-024-02947-5

Weitere Artikel der Ausgabe 1/2024

BMC Pulmonary Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

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.