Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Am J Respir Crit Care Med ; 205(9): 1084-1092, 2022 05 01.
Article in English | MEDLINE | ID: covidwho-1832814

ABSTRACT

Rationale: Chronic cough remains a major and often debilitating symptom for patients with idiopathic pulmonary fibrosis (IPF). In a phase 2A study, inhaled RVT-1601 (cromolyn sodium) reduced daytime cough and 24-hour average cough counts in patients with IPF. Objectives: To determine the efficacy, safety, and optimal dose of inhaled RVT-1601 for the treatment of chronic cough in patients with IPF. Methods: In this multicenter, randomized, placebo-controlled phase 2B study, patients with IPF and chronic cough for ⩾8 weeks were randomized (1:1:1:1) to receive 10, 40, and 80 mg RVT-1601 three times daily or placebo for 12 weeks. The primary endpoint was change from baseline to end of treatment in log-transformed 24-hour cough count. Key secondary endpoints were change from baseline in cough severity and cough-specific quality of life. Safety was monitored throughout the study. Measurements and Main Results: The study was prematurely terminated owing to the impact of the coronavirus disease (COVID-19) pandemic. Overall, 108 patients (mean age 71.0 years, 62.9% males) received RVT-1601 10 mg (n = 29), 40 mg (n = 25), 80 mg (n = 27), or matching placebo (n = 27); 61.1% (n = 66) completed double-blind treatment. No statistically significant difference was observed in the least-square mean change from baseline in log-transformed 24-hour average cough count, cough severity, and cough-specific quality of life score between the RVT-1601 groups and the placebo group. The mean percentage change from baseline in 24-hour average cough count was 27.7% in the placebo group. Treatment was generally well tolerated. Conclusions: Treatment with inhaled RVT-1601 (10, 40, and 80 mg three times a day) did not provide benefit over placebo for the treatment of chronic cough in patients with IPF.


Subject(s)
COVID-19 , Idiopathic Pulmonary Fibrosis , Aged , Chronic Disease , Cough/complications , Cough/etiology , Double-Blind Method , Female , Humans , Idiopathic Pulmonary Fibrosis/complications , Idiopathic Pulmonary Fibrosis/drug therapy , Male , Quality of Life , Treatment Outcome
2.
J Infect Chemother ; 28(7): 1029-1032, 2022 Jul.
Article in English | MEDLINE | ID: covidwho-1783494

ABSTRACT

A recent study reported that patients with interstitial lung disease (ILD) are at increased risk of death from coronavirus disease 2019 (COVID-19). However, there are no studies on the outcome of COVID-19 patients with preexisting ILD treated with corticosteroids or antiviral drugs. We extracted 26 patients with preexisting ILD by medical records and HRCT pattern. Of 503 patients with COVID-19, we selected 52 patients as control matched for age and sex. Twenty out of the 26 ILD patients (76.9%) received corticosteroid therapy, and 23 patients (88.5%) also received antiviral treatment with remdesivir or favipiravir. Although no statistical difference was found, the proportion of severe patients in ILD group tended to be higher than in non-ILD group (23.1% vs. 42.3%; p = 0.114). Also, mortality rate in ILD group tended to be higher than in non-ILD patients (11.5% vs. 3.8%; p = 0.326). In univariate analysis to evaluate risk factors for severe condition, diagnosis of idiopathic pulmonary fibrosis, usual interstitial pneumonia pattern, and honeycomb lung were not risk factors of severe disease. Treatment with corticosteroids, antiviral drugs, and immunosuppressive agents may affect the outcome of COVID-19 patients with ILD.


Subject(s)
COVID-19 Drug Treatment , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Adrenal Cortex Hormones/therapeutic use , Anti-Inflammatory Agents , Antiviral Agents/therapeutic use , Humans , Idiopathic Pulmonary Fibrosis/complications , Lung , Lung Diseases, Interstitial/drug therapy , Retrospective Studies , Tomography, X-Ray Computed
3.
BMJ Case Rep ; 14(10)2021 Oct 19.
Article in English | MEDLINE | ID: covidwho-1476412

ABSTRACT

A 79-year-old woman presented with a week-long history of shortness of breath. She had a background of idiopathic pulmonary fibrosis (IPF) which was stable and had not required any antifibrotic treatment. A month prior to this presentation, she was admitted with COVID-19 pneumonia, with maximal oxygen requirement of 2 L, but was discharged without need for supplemental oxygen. On readmission, she was found to have severe, rapidly progressive pulmonary fibrosis. After all precipitating causes were ruled out, it was felt her recent COVID-19 infection was the exacerbating factor causing progression of pulmonary fibrosis. COVID-19 infection has been hypothesised to cause long term pulmonary fibrosis, but this is the first case highlighting COVID-19 infection as the causative agent exacerbating IPF.


Subject(s)
COVID-19 , Idiopathic Pulmonary Fibrosis , Aged , Disease Progression , Female , Humans , Idiopathic Pulmonary Fibrosis/complications , Lung , SARS-CoV-2
4.
Brief Bioinform ; 22(2): 1254-1266, 2021 03 22.
Article in English | MEDLINE | ID: covidwho-1343630

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is accountable for the cause of coronavirus disease (COVID-19) that causes a major threat to humanity. As the spread of the virus is probably getting out of control on every day, the epidemic is now crossing the most dreadful phase. Idiopathic pulmonary fibrosis (IPF) is a risk factor for COVID-19 as patients with long-term lung injuries are more likely to suffer in the severity of the infection. Transcriptomic analyses of SARS-CoV-2 infection and IPF patients in lung epithelium cell datasets were selected to identify the synergistic effect of SARS-CoV-2 to IPF patients. Common genes were identified to find shared pathways and drug targets for IPF patients with COVID-19 infections. Using several enterprising Bioinformatics tools, protein-protein interactions (PPIs) network was designed. Hub genes and essential modules were detected based on the PPIs network. TF-genes and miRNA interaction with common differentially expressed genes and the activity of TFs are also identified. Functional analysis was performed using gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathway and found some shared associations that may cause the increased mortality of IPF patients for the SARS-CoV-2 infections. Drug molecules for the IPF were also suggested for the SARS-CoV-2 infections.


Subject(s)
COVID-19/complications , Idiopathic Pulmonary Fibrosis/complications , SARS-CoV-2/genetics , COVID-19/genetics , COVID-19/virology , Datasets as Topic , Epithelial Cells/virology , Gene Ontology , Genes, Viral , Humans , Lung/cytology , Lung/virology , Transcriptome
5.
J Mol Med (Berl) ; 99(10): 1373-1384, 2021 10.
Article in English | MEDLINE | ID: covidwho-1309024

ABSTRACT

Pulmonary fibrosis is a chronic debilitating condition characterized by progressive deposition of connective tissue, leading to a steady restriction of lung elasticity, a decline in lung function, and a median survival of 4.5 years. The leading causes of pulmonary fibrosis are inhalation of foreign particles (such as silicosis and pneumoconiosis), infections (such as post COVID-19), autoimmune diseases (such as systemic autoimmune diseases of the connective tissue), and idiopathic pulmonary fibrosis. The therapeutics currently available for pulmonary fibrosis only modestly slow the progression of the disease. This review is centered on the interplay of damage-associated molecular pattern (DAMP) molecules, Toll-like receptor 4 (TLR4), and inflammatory cytokines (such as TNF-α, IL-1ß, and IL-17) as they contribute to the pathogenesis of pulmonary fibrosis, and the possible avenues to develop effective therapeutics that disrupt this interplay.


Subject(s)
Alarmins/metabolism , Cytokines/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Inflammation/metabolism , Toll-Like Receptor 4/metabolism , Animals , Humans , Idiopathic Pulmonary Fibrosis/complications , Idiopathic Pulmonary Fibrosis/therapy , Inflammation/complications , Models, Biological
6.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: covidwho-1180574

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), better known as COVID-19, has become a current threat to humanity. The second wave of the SARS-CoV-2 virus has hit many countries, and the confirmed COVID-19 cases are quickly spreading. Therefore, the epidemic is still passing the terrible stage. Having idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) are the risk factors of the COVID-19, but the molecular mechanisms that underlie IPF, COPD, and CVOID-19 are not well understood. Therefore, we implemented transcriptomic analysis to detect common pathways and molecular biomarkers in IPF, COPD, and COVID-19 that help understand the linkage of SARS-CoV-2 to the IPF and COPD patients. Here, three RNA-seq datasets (GSE147507, GSE52463, and GSE57148) from Gene Expression Omnibus (GEO) is employed to detect mutual differentially expressed genes (DEGs) for IPF, and COPD patients with the COVID-19 infection for finding shared pathways and candidate drugs. A total of 65 common DEGs among these three datasets were identified. Various combinatorial statistical methods and bioinformatics tools were used to build the protein-protein interaction (PPI) and then identified Hub genes and essential modules from this PPI network. Moreover, we performed functional analysis under ontologies terms and pathway analysis and found that IPF and COPD have some shared links to the progression of COVID-19 infection. Transcription factors-genes interaction, protein-drug interactions, and DEGs-miRNAs coregulatory network with common DEGs also identified on the datasets. We think that the candidate drugs obtained by this study might be helpful for effective therapeutic in COVID-19.


Subject(s)
COVID-19/complications , Computational Biology/methods , Idiopathic Pulmonary Fibrosis/complications , Pulmonary Disease, Chronic Obstructive/complications , Systems Biology/methods , Humans , Protein Interaction Maps , SARS-CoV-2/isolation & purification
7.
Part Fibre Toxicol ; 18(1): 11, 2021 03 11.
Article in English | MEDLINE | ID: covidwho-1133602

ABSTRACT

BACKGROUND: Air pollution exposure and idiopathic pulmonary fibrosis (IPF) cause a poor prognosis after SARS-CoV-2 infection, but the underlying mechanisms are not well explored. Angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) are the keys to the entry of SARS-CoV-2. We therefore hypothesized that air pollution exposure and IPF may increase the expression of ACE2 and TMPRSS2 in the lung alveolar region. We measured their expression levels in lung tissues of control non-IPF and IPF patients, and used murine animal models to study the deterioration of IPF caused by particulate matter (PM) and the molecular pathways involved in the expression of ACE2 and TMPRSS2. RESULTS: In non-IPF patients, cells expressing ACE2 and TMPRSS2 were limited to human alveolar cells. ACE2 and TMPRSS2 were largely upregulated in IPF patients, and were co-expressed by fibroblast specific protein 1 (FSP-1) + lung fibroblasts in human pulmonary fibrotic tissue. In animal models, PM exposure increased the severity of bleomycin-induced pulmonary fibrosis. ACE2 and TMPRSS2 were also expressed in FSP-1+ lung fibroblasts in bleomycin-induced pulmonary fibrosis, and when combined with PM exposure, they were further upregulated. The severity of pulmonary fibrosis and the expression of ACE2 and TMPRSS2 caused by PM exposure were blocked by deletion of KC, a murine homologue of IL-8, or treatment with reparixin, an inhibitor of IL-8 receptors CXCR1/2. CONCLUSIONS: These data suggested that risk of SARS-CoV-2 infection and COVID-19 disease severity increased by air pollution exposure and underlying IPF. It can be mediated through upregulating ACE2 and TMPRSS2 in pulmonary fibroblasts, and prevented by blocking the IL-8/CXCR1/2 pathway.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/etiology , Idiopathic Pulmonary Fibrosis/complications , Particulate Matter/toxicity , SARS-CoV-2 , Serine Endopeptidases/genetics , Angiotensin-Converting Enzyme 2/physiology , Animals , Humans , Interleukin-8/physiology , Male , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/enzymology , Serine Endopeptidases/physiology , Up-Regulation
8.
Tuberk Toraks ; 68(4): 419-429, 2020 Dec.
Article in Turkish | MEDLINE | ID: covidwho-1067902

ABSTRACT

Though it has been 8 months since the beginning of COVID-19 pandemic, number of cases and deaths are still seriously increasing. We still don't have enough evidence about the prognosis of patients who had COVID-19 pneumonia. In long term follow up we wonder if they will have rapid FVC decline, widespread fibrosis in computed tomography, loss in quality of life or increased mortality that we experience in idiopathic pulmonary fibrosis, chronic hypersensitivity pneumonia or autoimmune interstitial lung diseases. However, in elderly patients less severe dysfunction or non-progressive-fibrosis can cause morbidity and mortality. Therefore, if we consider millions of people who are affected by COVID-19, even a rare complication can cause serious health problem in social scale. Because of the importance of this issue randomized controlled trials should be rapidly planned on post-COVID fibrosis, COVID associated thrombosis, risk factors, prevention and treatment (1). In this review, the frequency, clinical importance, prevention and treatment of possible long-term sequels of COVID-19 pneumonia (pulmonary fibrosis, pulmonary embolism and pulmonary hypertension) will be discussed.


Subject(s)
COVID-19/epidemiology , Idiopathic Pulmonary Fibrosis/prevention & control , Pandemics , SARS-CoV-2 , COVID-19/complications , Humans , Idiopathic Pulmonary Fibrosis/complications , Idiopathic Pulmonary Fibrosis/diagnostic imaging , Prognosis , Risk Factors , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL