1
|
Chen B, Leng Z, Zhang J, Shi X, Dong S, Wang B. Diagnostic Application of Bronchoalveolar Lavage Fluid Analysis in Cases of Idiopathic Pulmonary Fibrosis in which Diagnosis Cannot Be Confirmed by High-Resolution Computed Tomography. Lung 2025; 203:16. [PMID: 39751999 DOI: 10.1007/s00408-024-00758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/25/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive fibrotic lung disorder characterized by dry cough, fatigue, and exacerbated dyspnea. The prognosis of IPF is notably unfavorable, becoming extremely poor when the disease advances acutely. Effective therapeutic intervention is essential to mitigate disease progression; hence, early diagnosis and treatment are paramount. When high-resolution computed tomography (HRCT) reveals usual interstitial pneumonia (UIP), a diagnosis of IPF can be established. However, when HRCT fails to conclusively confirm IPF, the diagnostic pathway becomes intricate and necessitates a multidisciplinary approach involving clinicians, radiologists, and pathologists. Consequently, the objective of this study was to investigate new diagnostic approaches through bronchoalveolar lavage (BAL) analysis. METHODS BAL is a commonly utilized diagnostic tool for interstitial lung diseases. We review the application of bronchoalveolar lavage (BALF) in idiopathic pulmonary fibrotic disease, emphasizing that the cellular and solute composition of the lower respiratory tract offers valuable insights. RESULTS This review delineates the advancements in diagnosing IPF cases that remain indeterminate via HRCT, leveraging BALF analysis. In contrast to surgical lung biopsy, BAL is minimally invasive and offers potential diagnostic utility through the identification of specific BALF biomarkers. CONCLUSION Augment the clinical diagnostic armamentarium for IPF, particularly in scenarios where HRCT findings are inconclusive.
Collapse
Affiliation(s)
- Boyi Chen
- Department of Respiratory Medicine, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Zhefeng Leng
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
| | - Jianhui Zhang
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
| | - Xuefei Shi
- Department of Respiratory Medicine, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China.
| | - Shunli Dong
- Department of Respiratory Medicine, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China.
| | - Bin Wang
- Department of Respiratory Medicine, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China.
| |
Collapse
|
2
|
Krauss E, Tello S, Naumann J, Wobisch S, Ruppert C, Kuhn S, Mahavadi P, Majeed RW, Bonniaud P, Molina-Molina M, Wells A, Hirani N, Vancheri C, Walsh S, Griese M, Crestani B, Guenther A. Protocol and research program of the European registry and biobank for interstitial lung diseases (eurILDreg). BMC Pulm Med 2024; 24:572. [PMID: 39558302 PMCID: PMC11575435 DOI: 10.1186/s12890-024-03389-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND AND AIMS Interstitial lung diseases (ILDs), encompassing both pediatric and adult cases, present a diverse spectrum of chronic conditions with variable prognosis. Despite limited therapeutic options beyond antifibrotic drugs and immunosuppressants, accurate diagnosis is challenging, often necessitating invasive procedures that may not be feasible for certain patients. Drawn against this background, experts across pediatric and adult ILD fields have joined forces in the RARE-ILD initiative to pioneer novel non-invasive diagnostic algorithms and biomarkers. Collaborating with the RARE-ILD consortium, the eurILDreg aims to comprehensively describe different ILDs, analyze genetically defined forms across age groups, create innovative diagnostic and therapeutic biomarkers, and employ artificial intelligence for data analysis. METHODS The foundation of eurILDreg is built on a comprehensive parameter list developed and adopted by clinical experts, encompassing over 1,800 distinct parameters related to patient history, clinical examinations, diagnosis, lung function and biospecimen collection. This robust dataset is further enriched with daily assessments captured through the patientMpower app, including handheld spirometry and exercise tests, conducted on approximately 350 patients over the course of a year. This approach involves app-based daily assessments of quality of life, symptom tracking, handheld spirometry, saturation measurement, and the 1-min sit-to-stand test (1-STST). Additionally, pediatric data from the ChILD-EU registry will be integrated into the RARE-ILD Data Warehouse, with the ultimate goal of including a total of 4.000 ILD patients and over 100.000 biospecimen. DISCUSSION The collaborative efforts within the consortium are poised to streamline research endeavors significantly, promising to advance patient-centered care, foster innovation, and shape the future landscape of interstitial lung disease research and healthcare practices. TRIAL REGISTRATION EurILDreg is registered in the German Clinical Trials Register (DRKS 00028968, 26.07.2022), and eurIPFreg is registered in ClinicalTrials.gov (NCT02951416).
Collapse
Affiliation(s)
- Ekaterina Krauss
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Silke Tello
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Jennifer Naumann
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Sandra Wobisch
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Clemens Ruppert
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Stefan Kuhn
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Poornima Mahavadi
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
| | - Raphael W Majeed
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Klinikstr. 33, 35392, Giessen, Germany
| | - Philippe Bonniaud
- Service de Pneumologie Et Soins Intensifs Respiratoire, Centre de Référence Constitutif Des Maladies Pulmonaires Rares de L'Adultes de Dijon, Centre Hospitalier Universitaire de Dijon-Bourgogne, INSERM U1231, Equipe HSP-Pathies, Faculty of Medicine and Pharmacy, Université de Bourgogne, Dijon, France
| | - Maria Molina-Molina
- ILD Unit, Respiratory Department, University Hospital of Bellvitge (HUB), Biomedical Research Institute of Bellvitge (IDIBELL), Barcelona, Spain
| | | | - Nik Hirani
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, Regional Referral Center for Rare Lung Diseases, University Hospital Policlinico, University of Catania, Catania, Italy
| | - Simon Walsh
- King's College Hospital Foundation Trust, Denmark Hill, London, UK
| | - Matthias Griese
- ChILD-EU, Hauner Children's Hospital, University of Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Bruno Crestani
- Institute National de La Sainté Et de La Recherche Médicale, Hopital Bichat, Service de Pneumologie, Paris, France
| | - Andreas Guenther
- European IPF/ILD Registry & Biobank (eurIPFreg/Bank, eurILDreg/Bank), Giessen, Germany.
- Center for Interstitial and Rare Lung Diseases, Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Giessen, Germany.
- Cardio-Pulmonary Institute (CPI), Klinikstr. 33, 35392, Giessen, Germany.
- Agaplesion Lung Clinic "Evangelisches Krankenhaus Mittelhessen", Paul-Zipp Str. 171, 35398, Giessen, Germany.
- Institute for Lung Health (ILH), Giessen, Germany.
- eurILDreg Investigators, European ILD Registry (eurILDreg), Klinikstrasse 36, Giessen, 35392, Germany.
| |
Collapse
|
3
|
Gupta N, Paryani M, Patel S, Bariya A, Srivastava A, Pathak Y, Butani S. Therapeutic Strategies for Idiopathic Pulmonary Fibrosis - Thriving Present and Promising Tomorrow. J Clin Pharmacol 2024; 64:779-798. [PMID: 38346921 DOI: 10.1002/jcph.2408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/11/2024] [Indexed: 06/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a continuous, progressive, and lethal age-related respiratory disease. It is characterized by condensed and rigid lung tissue, which leads to a decline in the normal functioning of the lungs. The pathophysiology of IPF has still not been completely elucidated, so current strategies are lagging behind with respect to improving the condition of patients with IPF and increasing their survival rate. The desire for a better understanding of the pathobiology of IPF and its early detection has led to the identification of various biomarkers associated with IPF. The use of drugs such as pirfenidone and nintedanib as a safe and effective treatment alternative have marked a new chapter in the treatment of IPF. However, nonpharmacological therapies, involving long-term oxygen therapy, transplantation of the lungs, pulmonary rehabilitation, ventilation, and palliative care for cough and dyspnea, are still considered to be beneficial as supplementary methods for IPF therapy. A major risk factor for IPF is aging, with associated hallmarks such as telomere attrition, senescence, epigenetic drift, stem cell exhaustion, loss of proteostasis, and mitochondrial dysfunction. These are promising earmarks for the development of potential therapy for the disease. In this review, we have discussed current and emerging novel therapeutic strategies for IPF, especially for targets associated with age-related mechanisms.
Collapse
Affiliation(s)
- Nikita Gupta
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Mitali Paryani
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Snehal Patel
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Aditi Bariya
- Arihant School of Pharmacy Education and Research, Adalaj, Gandhinagar, Gujarat, India
| | - Anshu Srivastava
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Yashwant Pathak
- USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, USA
- Faculty of Pharmacy, Airlangga University, Surabaya, Indonesia
| | - Shital Butani
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
4
|
Okaba K, Inokuchi G, Horioka K, Iwase H, Inoue H, Motomura A, Ishii N, Moue C, Shiomi T, Yajima D. Forensic application of three interstitial pneumonia markers: search for new pneumonia markers in dead bodies. Int J Legal Med 2024; 138:1583-1592. [PMID: 38379061 DOI: 10.1007/s00414-024-03187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 02/09/2024] [Indexed: 02/22/2024]
Abstract
In forensic cases, detailed identification of pneumonia is important. Our objective was to statistically determine the applicability of three interstitial lung disease (ILD) markers for forensic diagnosis using serum collected from dead bodies with various postmortem intervals (PMIs). We retrospectively analyzed the levels of postmortem serum Krebs von den Lungen-6 (KL-6) and pulmonary surfactant-associated proteins A and D (SP-A and SP-D) using 221 samples obtained during forensic autopsy at our facility from 2019 to 2023. We evaluated the diagnostic efficacy of ILD markers for various pneumonias against the pathological diagnosis, and examined the assessment of the severity of ILD. When comparing the ILD group with bacterial pneumonia (BP) versus the control group, there was a significant increase in KL-6 in the ILD group. When comparing the severe ILD (SILD) group with the mild ILD (MILD) group, there was a significant increase in KL-6 and SP-D in the SILD group. The optimal cutoff values for differentiating SILD were 607.0 U/mL for KL-6, 55.5 ng/mL for SP-A, and 160.0 ng/mL for SP-D, and the sensitivity/specificity (%) of KL-6, SP-A, and SP-D for SILD were 84.1/95.2, 55.6/85.7, and 66.7/74.6, respectively. This is the first study to examine KL-6 in postmortem serum in forensic medicine. By analyzing dead bodies with various PMIs, our results confirmed statistically that postmortem serum KL-6 specifically detects ILD, postmortem serum SP-A has high sensitivity to lung injury, and postmortem serum SP-D is potentially useful in assessing the severity of ILD.
Collapse
Affiliation(s)
- Keisuke Okaba
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan.
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Go Inokuchi
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kie Horioka
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
- Department of Forensic Medicine, Research Unit of Internal Medicine, Medical Research Center, University of Oulu, Oulu, Finland
| | - Hirotaro Iwase
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroyuki Inoue
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Ayumi Motomura
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Namiko Ishii
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Chihiro Moue
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Takayuki Shiomi
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Daisuke Yajima
- Department of Forensic Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
- Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
5
|
Simón-Fuentes M, Ríos I, Herrero C, Lasala F, Labiod N, Luczkowiak J, Roy-Vallejo E, Fernández de Córdoba-Oñate S, Delgado-Wicke P, Bustos M, Fernández-Ruiz E, Colmenares M, Puig-Kröger A, Delgado R, Vega MA, Corbí ÁL, Domínguez-Soto Á. MAFB shapes human monocyte-derived macrophage response to SARS-CoV-2 and controls severe COVID-19 biomarker expression. JCI Insight 2023; 8:e172862. [PMID: 37917179 PMCID: PMC10807725 DOI: 10.1172/jci.insight.172862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
Monocyte-derived macrophages, the major source of pathogenic macrophages in COVID-19, are oppositely instructed by macrophage CSF (M-CSF) or granulocyte macrophage CSF (GM-CSF), which promote the generation of antiinflammatory/immunosuppressive MAFB+ (M-MØ) or proinflammatory macrophages (GM-MØ), respectively. The transcriptional profile of prevailing macrophage subsets in severe COVID-19 led us to hypothesize that MAFB shapes the transcriptome of pulmonary macrophages driving severe COVID-19 pathogenesis. We have now assessed the role of MAFB in the response of monocyte-derived macrophages to SARS-CoV-2 through genetic and pharmacological approaches, and we demonstrate that MAFB regulated the expression of the genes that define pulmonary pathogenic macrophages in severe COVID-19. Indeed, SARS-CoV-2 potentiated the expression of MAFB and MAFB-regulated genes in M-MØ and GM-MØ, where MAFB upregulated the expression of profibrotic and neutrophil-attracting factors. Thus, MAFB determines the transcriptome and functions of the monocyte-derived macrophage subsets that underlie pulmonary pathogenesis in severe COVID-19 and controls the expression of potentially useful biomarkers for COVID-19 severity.
Collapse
Affiliation(s)
- Miriam Simón-Fuentes
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Israel Ríos
- Immunometabolism and Inflammation Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Cristina Herrero
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Fátima Lasala
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Nuria Labiod
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Emilia Roy-Vallejo
- Rheumatology Department, University Hospital La Princesa and Research Institute, Madrid, Spain
| | | | - Pablo Delgado-Wicke
- Molecular Biology Unit, University Hospital La Princesa and Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Matilde Bustos
- Institute of Biomedicine of Seville (IBiS), Spanish National Research Council (CSIC), University of Seville, Virgen del Rocio University Hospital (HUVR), Seville, Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit, University Hospital La Princesa and Research Institute, Universidad Autónoma de Madrid, Madrid, Spain
| | - Maria Colmenares
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Amaya Puig-Kröger
- Immunometabolism and Inflammation Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | | |
Collapse
|
6
|
Lederer C, Mayer K, Somogyi V, Kriegsmann K, Kriegsmann M, Buschulte K, Polke M, Findeisen P, Herth F, Kreuter M. Krebs von den Lungen-6 as a Potential Predictive Biomarker in Fibrosing Interstitial Lung Diseases. Respiration 2023; 102:591-600. [PMID: 37586349 DOI: 10.1159/000531945] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/11/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND As fibrosing interstitial lung diseases (fILDs) are associated with high mortality, monitoring of disease activity under treatment is highly relevant. Krebs von den Lungen-6 (KL-6) is associated with the presence and severity of different fILDs, mainly in Asian patient populations. OBJECTIVES Our aim was to evaluate KL-6 as a predictive biomarker in fILDs in Caucasian patients. METHODS Consecutive patients with fILDs were recruited prospectively and serum concentrations of KL-6 were measured at baseline (BL), after 6 and 12 months (6 Months, 12 Months). Clinical characteristics including pulmonary function tests were assessed at 6-monthly visits and correlated with KL-6 BL levels as well as with KL-6 level changes. RESULTS A total of 47 fILD patients were included (mean age: 65 years, 68% male). KL-6 levels at BL were significantly higher in fILD patients than in healthy controls (n = 44, mean age: 45, 23% male) (ILD: 1,757 ± 1960 U/mL vs. control: 265 ± 107 U/mL, p < 0.0001). However, no differences were noted between ILD subgroups. KL-6 decreased significantly under therapy (6M∆BL-KL6: -486 ± 1,505 mean U/mL, p = 0.032; 12M∆BL-KL6: -547 ± 1,782 mean U/mL, p = 0.041) and KL-6 level changes were negatively correlated with changes in pulmonary function parameters (forced vital capacity [FVC]: r = -0.562, p < 0.0001; DLCOSB: r = -0.405, p = 0.013). While neither absolute KL-6 levels at BL nor KL-6 level changes were associated with ILD progression (FVC decline ≥10%, DLCOSB decline ≥15% or death), patients with a stable FVC showed significantly decreasing KL-6 levels (p = 0.022). CONCLUSIONS A decline of KL-6 under therapy correlated with a clinically relevant stabilization of lung function. Thus, KL-6 might serve as a predictive biomarker, which however must be determined by larger prospective cohorts.
Collapse
Affiliation(s)
- Christoph Lederer
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany,
| | - Katharina Mayer
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
| | - Vivien Somogyi
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
- Mainz Center for Pulmonary Medicine, Department of Pneumology, Mainz University Medical Center and Department of Pulmonary, Critical Care & Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidleberg, Heidelberg, Germany
| | - Mark Kriegsmann
- Department of Pathology, University Hospital Heidelberg, Germany and Pathology Wiesbaden, Wiesbaden, Germany
| | - Katharina Buschulte
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
| | - Markus Polke
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
| | | | - Felix Herth
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Pneumology, Thoraxklinik Heidelberg, University Hospital Heidelberg and German Center for Lung Research (DZL), Heidelberg, Germany
- Mainz Center for Pulmonary Medicine, Department of Pneumology, Mainz University Medical Center and Department of Pulmonary, Critical Care & Sleep Medicine, Marienhaus Clinic Mainz, Mainz, Germany
| |
Collapse
|
7
|
Rai M, Parthasarathi A, Beeraka NM, Kaleem Ullah M, Malamardi S, Padukudru S, Siddaiah JB, Uthaiah CA, Vishwanath P, Chaya SK, Ramaswamy S, Upadhyay S, Ganguly K, Mahesh PA. Circulatory Serum Krebs von Den Lungen-6 and Surfactant Protein-D Concentrations Predict Interstitial Lung Disease Progression and Mortality. Cells 2023; 12:cells12091281. [PMID: 37174681 PMCID: PMC10177381 DOI: 10.3390/cells12091281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
There is a need for biomarkers to predict outcomes, including mortality, in interstitial lung disease (ILD). Krebs von den Lungen-6 (KL-6) and surfactant protein D (SP-D) are associated with lung damage and fibrosis in all ILDs and are related to important clinical outcomes. Though these two biomarkers have been associated with ILD outcomes, there are no studies that have evaluated their predictive potential in combination. This study aims to determine whether KL-6 and SP-D are linked to poor disease outcomes and mortality. Additionally, we plan to examine whether changes in KL-6 and SP-D concentrations correspond with changes in lung function and whether serial measurements improve their predictive potential to identify disease progression and mortality. Forty-four patients with ILD participated in a prospective 6-month longitudinal observational study. ILD patients who succumbed had the highest KL-6 levels (3990.4 U/mL (3490.0-4467.6)) and highest SP-D levels (256.1 ng/mL (217.9-260.0)), followed by those who deteriorated: KL-6 levels 1357.0 U/mL (822.6-1543.4) and SP-D levels 191.2 ng/mL (152.8-210.5). The generalized linear model (GLM) analysis demonstrated that changes in forced vital capacity (FVC), diffusing capacity of lungs for carbon monoxide (DLCO), forced expiratory volume in 1 s (FEV1), and partial pressure of arterial oxygen (PaO2) were correlated to changes in KL6 (p = 0.016, 0.014, 0.027, 0.047) and SP-D (p = 0.008, 0.012, 0.046, 0.020), respectively. KL-6 (odds ratio (OR): 2.87 (1.06-7.79)) and SPD (OR: 1.76 (1.05-2.97)) were independent predictors of disease progression, and KL-6 (hazard ratio (HR): 3.70 (1.46-9.41)) and SPD (HR: 2.58 (1.01-6.59)) were independent predictors of death by Cox regression analysis. Combined biomarkers (KL6 + SPD + CT + FVC) had the strongest ability to predict disease progression (AUC: 0.797) and death (AUC: 0.961), on ROC analysis. Elevated KL-6 and SPD levels are vital biomarkers for predicting the severity, progression, and outcomes of ILD. High baseline levels or an increase in levels over a six-month follow-up despite treatment indicate a poor prognosis. Combining KL6 and SPD with conventional measures yields a more potent prognostic indicator. Clinical studies are needed to test additional interventions, and future research will determine if this combined biomarker benefits different ethnicities globally.
Collapse
Affiliation(s)
- Meghna Rai
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Ashwaghosha Parthasarathi
- Allergy, Asthma, and Chest Centre, Krishnamurthypuram, Mysuru 570004, India
- Rutgers Centre for Pharmacoepidemiology and Treatment Science, New Brunswick, NJ 08901-1293, USA
| | - Narasimha M Beeraka
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu 515721, Andhra Pradesh, India
| | - Mohammed Kaleem Ullah
- Centre for Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research, Mysore 570015, India
- Division of Infectious Disease and Vaccinology, School of Public Health, University of California, Berkeley, CA 94720, USA
| | - Sowmya Malamardi
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
- School of Psychology & Public Health, College of Science Health and Engineering, La Trobe University, Melbourne 3086, Australia
| | - Sunag Padukudru
- Yenepoya Medical College, Yenepoya University, Mangalore 575018, Karnataka, India
| | - Jayaraj Biligere Siddaiah
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Chinnappa A Uthaiah
- Centre for Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Prashant Vishwanath
- Centre for Excellence in Molecular Biology and Regenerative Medicine (A DST-FIST Supported Center), Department of Biochemistry (A DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research, Mysore 570015, India
| | - Sindaghatta Krishnarao Chaya
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Subramanian Ramaswamy
- Department of Clinical Immunology & Rheumatology, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
| | - Swapna Upadhyay
- Unit of Integrative Toxicology, Institute of Environmental Medicine (IMM), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine (IMM), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Padukudru Anand Mahesh
- Department of Respiratory Medicine, JSS Medical College, JSS Academy of Higher Education and Research, Mysuru 570015, India
| |
Collapse
|
8
|
Kang D, Lee Y, Kim W, Lee HR, Jung S. 3D pulmonary fibrosis model for anti-fibrotic drug discovery by inkjet-bioprinting. Biomed Mater 2022; 18. [PMID: 36562496 DOI: 10.1088/1748-605x/aca8e3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis (PF) is known as a chronic and irreversible disease characterized by excessive extracellular matrix accumulation and lung architecture changes. Large efforts have been made to develop prospective treatments and study the etiology of pulmonary fibrotic diseases utilizing animal models and spherical organoids. As part of these efforts, we created an all-inkjet-printed three-dimensional (3D) alveolar barrier model that can be used for anti-fibrotic drug discovery. Then, we developed a PF model by treating the 3D alveolar barrier with pro-fibrotic cytokine and confirmed that it is suitable for the fibrosis model by observing changes in structural deposition, pulmonary function, epithelial-mesenchymal transition, and fibrosis markers. The model was tested with two approved anti-fibrotic drugs, and we could observe that the symptoms in the disease model were alleviated. Consequently, structural abnormalities and changes in mRNA expression were found in the induced fibrosis model, which were shown to be recovered in all drug treatment groups. The all-inkjet-printed alveolar barrier model was reproducible for disease onset and therapeutic effects in the human body. This finding emphasized that thein vitroartificial tissue with faithfully implemented 3D microstructures using bioprinting technology may be employed as a novel testing platform and disease model to evaluate potential drug efficacy.
Collapse
Affiliation(s)
- Dayoon Kang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Yunji Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Wookyeom Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Hwa-Rim Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| | - Sungjune Jung
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea.,Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang 37673, Republic of Korea
| |
Collapse
|
9
|
Li X, Liu Y, Cheng L, Huang Y, Yan S, Li H, Zhan H, Li Y. Roles of biomarkers in anti-MDA5-positive dermatomyositis, associated interstitial lung disease, and rapidly progressive interstitial lung disease. J Clin Lab Anal 2022; 36:e24726. [PMID: 36221983 DOI: 10.1002/jcla.24726] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/12/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Anti-melanoma differentiation-associated gene 5 (MDA5)-positive dermatomyositis (MDA5+ DM) is significantly associated with interstitial lung disease (ILD), especially rapidly progressive ILD (RPILD) due to poor prognosis, resulting in high mortality rates. However, the pathogenic mechanism of MDA5+ DM-RPILD is unclear. Although some MDA5+ DM patients have a chronic course of ILD, many do not develop RPILD. Therefore, the related biomarkers for the early diagnosis, disease activity monitoring, and prediction of the outcome of RPILD in MDA5+ DM patients should be identified. Blood-based biomarkers are minimally invasive and can be easily detected. METHODS Recent relative studies related to blood biomarkers in PubMed were reviewed. RESULTS An increasing number of studies have demonstrated that dysregulated expression of blood biomarkers related to ILD such as ferritin, Krebs von den Lungen-6 (KL-6), surfactant protein-D (SP-D), and cytokines, and some tumor markers in MDA5+ DM may provide information in disease presence, activity, treatment response, and prognosis. These studies have highlighted the great potentials of blood biomarker values for MDA5+ DM-ILD and MDA5+ DM-RPILD. This review provides an overview of recent studies related to blood biomarkers, besides highlighted protein biomarkers, including antibody (anti-MDA5 IgG subclasses and anti-Ro52 antibody), genetic (exosomal microRNAs and neutrophil extracellular traps related to cell-free DNA), and immune cellular biomarkers in MDA5+ DM, MDA5+ DM-ILD, and MDA5+ DM-RPILD patients, hopefully elucidating the pathogenesis of MDA5+ DM-ILD and providing information on the early diagnosis, disease activity monitoring, and prediction of the outcome of the ILD, especially RPILD. CONCLUSIONS Therefore, this review may provide insight to guide treatment decisions for MDA5+ DM-RPILD patients and improve outcomes.
Collapse
Affiliation(s)
- Xiaomeng Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongmei Liu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Songxin Yan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Patel H, Shah JR, Patel DR, Avanthika C, Jhaveri S, Gor K. Idiopathic pulmonary fibrosis: Diagnosis, biomarkers and newer treatment protocols. Dis Mon 2022:101484. [DOI: 10.1016/j.disamonth.2022.101484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
11
|
Wu X, Jeong Y, Poli de Frías S, Easthausen I, Hoffman K, Oromendia C, Taheri S, Esposito AJ, Quesada Arias L, Ayaub EA, Maurer R, Gill RR, Hatabu H, Nishino M, Frits ML, Iannaccone CK, Weinblatt ME, Shadick NA, Dellaripa PF, Choi AMK, Kim EY, Rosas IO, Martinez FJ, Doyle TJ. Serum proteomic profiling of rheumatoid arthritis-interstitial lung disease with a comparison to idiopathic pulmonary fibrosis. Thorax 2022; 77:1041-1044. [PMID: 35907639 PMCID: PMC9976796 DOI: 10.1136/thorax-2021-217822] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 06/13/2022] [Indexed: 11/03/2022]
Abstract
Although interstitial lung disease (ILD) causes significant morbidity and mortality in rheumatoid arthritis (RA), it is difficult to predict the development or progression of ILD, emphasising the need for improved discovery through minimally invasive diagnostic tests. Aptamer-based proteomic profiling was used to assess 1321 proteins from 159 patients with rheumatoid arthritis with interstitial lung disease (RA-ILD), RA without ILD, idiopathic pulmonary fibrosis and healthy controls. Differential expression and gene set enrichment analyses revealed molecular signatures that are strongly associated with the presence and severity of RA-ILD and provided insight into unexplored pathways of disease. These warrant further study as non-invasive diagnostic tools and future therapeutic targets.
Collapse
Affiliation(s)
- Xiaoping Wu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Yunju Jeong
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | - Imaani Easthausen
- Department of Population Health Science, Division of Biostatistics, Weill Cornell Medicine, New York, New York, USA
| | - Katherine Hoffman
- Department of Population Health Science, Division of Biostatistics, Weill Cornell Medicine, New York, New York, USA
| | - Clara Oromendia
- Department of Population Health Science, Division of Biostatistics, Weill Cornell Medicine, New York, New York, USA
| | - Shahrad Taheri
- Department of Medicine, Weill Cornell Medicine-Qatar, Ar-Rayyan, Qatar
| | - Anthony J Esposito
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Luisa Quesada Arias
- Department of Medicine, Mount Sinai Medical Center, Miami Beach, Florida, USA
| | - Ehab A Ayaub
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Rie Maurer
- Center for Clinical Investigation, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ritu R Gill
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Hiroto Hatabu
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Mizuki Nishino
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michelle L Frits
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Christine K Iannaccone
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Michael E Weinblatt
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Nancy A Shadick
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Paul F Dellaripa
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Augustine M K Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Edy Y Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan O Rosas
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Fernando J Martinez
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Tracy J Doyle
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Kuronuma K, Otsuka M, Wakabayashi M, Yoshioka T, Kobayashi T, Kameda M, Morioka Y, Chiba H, Takahashi H. Role of transient receptor potential vanilloid 4 in therapeutic anti-fibrotic effects of pirfenidone. Am J Physiol Lung Cell Mol Physiol 2022; 323:L193-L205. [PMID: 35787697 DOI: 10.1152/ajplung.00565.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fatal lung disorder characterized by aberrant extracellular matrix deposition in the interstitium. Pirfenidone is an anti-fibrotic agent used to treat patients with IPF. Pirfenidone shows a pleiotropic mode of action, but its underlying anti-fibrotic mechanism is unclear. Transient receptor potential vanilloid 4 (TRPV4), which is a mechanosensitive calcium channel, was recently shown to be related to pulmonary fibrosis. To clarify the anti-fibrotic mechanisms of pirfenidone, we investigated whether TRPV4 blockade has a pharmacological effect in a murine model of pulmonary fibrosis and whether pirfenidone contributes to suppression of TRPV4. Our synthetic TRPV4 antagonist and pirfenidone treatment attenuated lung injury in the bleomycin mouse model. TRPV4-mediated increases in intracellular calcium were inhibited by pirfenidone. Additionally, TRPV4-stimulated interleukin-8 release from cells was reduced and a delay in cell migration was abolished by pirfenidone. Furthermore, pirfenidone decreased TRPV4 endogenous ligands in bleomycin-administered mouse lungs and their production by microsomes of human lungs. We found TRPV4 expression in the bronchiolar and alveolar epithelium and activated fibroblasts of the lungs in patients with IPF. Finally, we showed that changes in forced vital capacity of patients with IPF treated with pirfenidone were significantly correlated with metabolite levels of TRPV4 endogenous ligands in bronchoalveolar lavage fluid. These results suggest that the anti-fibrotic action of pirfenidone is partly mediated by TRPV4 and that TRPV4 endogenous ligands in bronchoalveolar lavage fluid may be biomarkers for distinguishing responders to pirfenidone.
Collapse
Affiliation(s)
- Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine, Sapporo-Kosei General Hospital, Sapporo, Japan
| | - Masato Wakabayashi
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Takeshi Yoshioka
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Tomofumi Kobayashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masami Kameda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuhide Morioka
- Drug Discovery and Disease Research Laboratory, Shionogi Co., Ltd., Osaka, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
13
|
Lin L, Zhao Y, Li Z, Li Y, Wang W, Kang J, Wang Q. Expression of S100A9 and KL-6 in common interstitial lung diseases. Medicine (Baltimore) 2022; 101:e29198. [PMID: 35512076 PMCID: PMC9276110 DOI: 10.1097/md.0000000000029198] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/10/2022] [Indexed: 01/04/2023] Open
Abstract
By evaluating S100 calcium binding protein A9 (S100A9) and Klebs von den Lungen-6 (KL-6) expression in patients with 4 common interstitial lung diseases (ILDs), we aimed to investigate whether S100A9 or KL-6 can be of any value in the differential diagnosis of these ILDs and simultaneously signal the disease progression.We collected the data of patients diagnosed with the 4 ILDs and underwent fiber-optic bronchoscopy and BAL in the First Affiliated Hospital, China Medical University from January 2012 to December 2020. The data related to BGA, C-reactive protein, pulmonary function test, total number and fraction of cells, T lymphocyte subsets in bronchoalveolar lavage fluid (BALF), and the expression of S100A9 and KL-6 in BALF and serum were collected. We analyzed, whether S100A9 or KL-6 could serve as a biomarker for differential diagnosis between the 4 common ILDs; whether the levels of S100A9 and KL-6 correlated with each other; whether they were correlated with other clinical parameters and disease severity.This study included 98 patients, 37 patients with idiopathic pulmonary fibrosis (IPF), 12 with hypersensitivity pneumonitis, 13 with connective tissue disease-associated ILD, and 36 with sarcoidosis (SAR): stage I (18), stage II (9), stage III (5), and stage IV (4). The expression of KL-6 in BALF was significantly higher in IPF patients than other 3 groups (all P-value < .05). However, there was no significant difference in the levels of S100A9 in BALF and serum between the 4 groups (P-value > .05). The levels of S100A9 in BALF of IPF patients was positively and significantly correlated with KL-6 expression and the percentage of neutrophils in BALF (P-value < .05). Along with the stage increase of SAR patients, the level of S100A9 in BALF gradually increased, which was negatively and significantly correlated with the forced vital capacity/predicted, carbon monoxide diffusing capacity/predicted%, and PaO2 (all P-value < .05).The expression of KL-6 in BALF can be used as a biomarker to differentiate IPF from the other 3 common ILDs. While, this was not the case with expression of S100A9 in BALF and serum. However, the expression S100A9 in BALF is useful to indicate the progression of SAR. Thus, simultaneous measurement of KL-6 and S100A9 levels in BALF makes more sense in differential diagnosing of the 4 common ILDS.
Collapse
Affiliation(s)
- Li Lin
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| | - Yabin Zhao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| | - Zhenhua Li
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| | - Yun Li
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
- Department of Geriatric Respiratory, The First Hospital of Kunming Medical University, Kunming, China
| | - Wei Wang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| | - Qiuyue Wang
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
14
|
Khan FA, Stewart I, Saini G, Robinson KA, Jenkins RG. A systematic review of blood biomarkers with individual participant data meta-analysis of matrix metalloproteinase-7 in idiopathic pulmonary fibrosis. Eur Respir J 2022; 59:2101612. [PMID: 34588192 PMCID: PMC9202487 DOI: 10.1183/13993003.01612-2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/21/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Blood-derived biomarkers have been described extensively as potential prognostic markers in idiopathic pulmonary fibrosis (IPF), but studies have been limited by analyses using data-dependent thresholds, inconsistent adjustment for confounders and an array of end-points, thus often yielding ungeneralisable results. Meta-analysis of individual participant data (IPD) is a powerful tool to overcome these limitations. Through systematic review of blood-derived biomarkers, sufficient studies with measurements of matrix metalloproteinase (MMP)-7 were identified to facilitate standardised analyses of the prognostic potential of this biomarker in IPF. METHODS Electronic databases were searched on 12 November 2020 to identify prospective studies reporting outcomes in patients with untreated IPF, stratified according to at least one pre-specified biomarker, measured at either baseline, or change over 3 months. IPD were sought for studies investigating MMP-7 as a prognostic factor. The primary outcome was overall mortality according to standardised MMP-7 z-scores, with a secondary outcome of disease progression in 12 months, all adjusted for age, gender, smoking and baseline forced vital capacity. RESULTS IPD was available for nine studies out of 12 identified, reporting outcomes from 1664 participants. Baseline MMP-7 levels were associated with increased mortality risk (adjusted hazard ratio 1.23, 95% CI 1.03-1.48; I2=64.3%) and disease progression (adjusted OR 1.27, 95% CI 1.11-1.46; I2=5.9%). In limited studies, 3-month change in MMP-7 was not associated with outcomes. CONCLUSION IPD meta-analysis demonstrated that greater baseline MMP-7 levels were independently associated with an increased risk of poor outcomes in patients with untreated IPF, while short-term changes did not reflect disease progression.
Collapse
Affiliation(s)
- Fasihul A Khan
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, UK
| | - Iain Stewart
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Health and Lung Institute, Imperial College London, London, UK
| | - Gauri Saini
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Karen A Robinson
- Epidemiology and Health Policy and Management, Johns Hopkins University, Baltimore, MD, USA
| | - R Gisli Jenkins
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
- Nottingham Biomedical Research Centre, National Institute for Health Research, Nottingham, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, National Health and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
15
|
Inhibition of lung microbiota-derived proapoptotic peptides ameliorates acute exacerbation of pulmonary fibrosis. Nat Commun 2022; 13:1558. [PMID: 35322016 PMCID: PMC8943153 DOI: 10.1038/s41467-022-29064-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 02/21/2022] [Indexed: 11/08/2022] Open
Abstract
Idiopathic pulmonary fibrosis is an incurable disease of unknown etiology. Acute exacerbation of idiopathic pulmonary fibrosis is associated with high mortality. Excessive apoptosis of lung epithelial cells occurs in pulmonary fibrosis acute exacerbation. We recently identified corisin, a proapoptotic peptide that triggers acute exacerbation of pulmonary fibrosis. Here, we provide insights into the mechanism underlying the processing and release of corisin. Furthermore, we demonstrate that an anticorisin monoclonal antibody ameliorates lung fibrosis by significantly inhibiting acute exacerbation in the human transforming growth factorβ1 model and acute lung injury in the bleomycin model. By investigating the impact of the anticorisin monoclonal antibody in a general model of acute lung injury, we further unravel the potential of corisin to impact such diseases. These results underscore the role of corisin in the pathogenesis of acute exacerbation of pulmonary fibrosis and acute lung injury and provide a novel approach to treating this incurable disease.
Collapse
|
16
|
Zhang Q, Wang Y, Tian C, Yu J, Li Y, Yang J. Clinical characteristics and genetic analysis of a Chinese pedigree of type 2 diabetes complicated with interstitial lung disease. Front Endocrinol (Lausanne) 2022; 13:1050200. [PMID: 36733806 PMCID: PMC9887333 DOI: 10.3389/fendo.2022.1050200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Diabetes mellitus is a systemic metabolic disorder which may target the lungs and lead to interstitial lung disease. The clinical characteristics and mechanisms of type 2 diabetes mellitus (T2DM) complicated with interstitial lung disease (ILD) have been studied. However, little work has been done to assess genetic contributions to the development of T2DM complicated with ILD. METHOD A pedigree of T2DM complicated with ILD was investigated, and the whole genome re-sequencing was performed to identify the genetic variations in the pedigree. According to the literature, the most valuable genetic contributors to the pathogenesis of T2DM complicated with ILD were screened out, and the related cellular functional experiments were also performed. RESULTS A large number of SNPs, InDels, SVs and CNVs were identified in eight subjects including two diabetic patients with ILD, two diabetic patients without ILD, and four healthy subjects from the pedigree. After data analysis according to the literature, MUC5B SNP rs2943512 (A > C) was considered to be an important potentially pathogenic gene mutation associated with the pathogenesis of ILD in T2DM patients. In vitro experiments showed that the expression of MUC5B in BEAS-2B cells was significantly up-regulated by high glucose stimulation, accompanied by the activation of ERK1/2 and the increase of IL-1β and IL-6. When silencing MUC5B by RNA interference, the levels of p-ERK1/2 as well as IL-1β and IL-6 in BEAS-2B cells were all significantly decreased. CONCLUSION The identification of these genetic variants in the pedigree enriches our understanding of the potential genetic contributions to T2DM complicated with ILD. MUC5B SNP rs2943512 (A > C) or the up-regulated MUC5B in bronchial epithelial cells may be an important factor in promoting ILD inT2DM patients, laying a foundation for future exploration about the pathogenesis of T2DM complicated with ILD.
Collapse
Affiliation(s)
- Qinghua Zhang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Chang Tian
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Jinyan Yu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanlei Li
- Department of Laboratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Junling Yang,
| |
Collapse
|
17
|
The Prognostic Value of Krebs von den Lungen-6 and Surfactant Protein-A Levels in the Patients with Interstitial Lung Disease. J Transl Int Med 2021; 9:212-222. [PMID: 34900632 PMCID: PMC8629416 DOI: 10.2478/jtim-2021-0040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background and Objectives The highly variable clinical course of interstitial lung disease (ILD) makes it difficult to predict patient prognosis. Serum surfactant protein-A (SP-A) and Krebs von den Lungen-6 (KL-6) are known prognostic biomarkers. However, the clinical or pathophysiological differences in patients with these biomarkers have not been well evaluated. We investigated the clinical and pathophysiological differences through the comparison of SP-A and KL-6 levels before and after treatment. Methods This study included retrospective data from 91 patients who were treated for ILD between August 2015 and September 2019. Serum SP-A and KL-6 levels were measured before and after treatment. The patients were followed up for 3 months. Results Changes in the serum biomarkers (Delta SP-A and Delta KL-6) were found to be significantly correlated (rs = 0.523, P < 0.001); Delta SP-A and Delta KL-6 were inversely correlated with changes in pulmonary function (% predicted values of diffusing capacity for carbon monoxide [DLCO], forced vital capacity [FVC], and forced expiratory volume in 1 s [FEV1]). Patients were divided into four groups based on their Delta SP-A and Delta KL-6 levels in a cluster analysis (G1, G2, G3, and G4). Both SP-A and KL-6 were elevated in the G1 group, with all the patients enrolled classified as progressive or unchanged, and 86.4% of patients showed improved disease activity in the G4 group, where both SP-A and KL-6 levels were reduced. In the G2 group, only SP-A levels decreased post-treatment, indicating an improvement in respiratory function; the patients were not at the end stage of the disease. Only the SP-A levels increased in the G3 group with immunosuppressive treatment. Conclusions Reduced serum SP-A and/or KL-6 levels are associated with improved lung function in patients with ILD. Some patients only showed a decrease in SP-A levels could prognosis an improvement in respiratory function. When only SP-A is increased, it may imply that the patients are at an early stage of disease progression. As a result, for proper disease monitoring, measuring both markers is important.
Collapse
|
18
|
Sakamoto N, Hamada N, Okamoto M, Tobino K, Ichiyasu H, Ishii H, Ichikado K, Morimoto S, Hosogaya N, Mukae H. Efficacy and safety of nintedanib in Japanese patients with early-stage idiopathic pulmonary fibrosis: a study protocol for an observational study. BMJ Open 2021; 11:e047249. [PMID: 34187824 PMCID: PMC8245432 DOI: 10.1136/bmjopen-2020-047249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION Idiopathic pulmonary fibrosis (IPF) is a fibrotic disease of unknown aetiology with a poor prognosis. Several clinical trials of nintedanib in patients with IPF have reported its inhibitory effect on reduced lung function, incidence of acute exacerbation of IPF and worsened health-related quality of life. Although nintedanib has a manageable safety and tolerability profile over long-term use, it was discontinued in over 20% of patients because of adverse events such as diarrhoea and liver dysfunction. This might explain why nintedanib use in patients with IPF is not widespread, especially among patients with early-stage IPF. In the present study, we aimed to clarify the efficacy, safety and tolerability of nintedanib in patients with stage I/II IPF, based on the Japanese IPF disease severity staging classification system. METHODS AND ANALYSIS This is an ongoing, prospective, multicentre observational cohort study of patients with stage I/II IPF who will start receiving nintedanib. Totally, 215 patients at 35 sites in Kyushu and Okinawa, Japan will be enrolled and followed up for 3 years. Nintedanib therapy would be initiated at the discretion of the investigator. The primary endpoint, change in forced vital capacity (FVC) at 156 weeks, will be shown as the mean change in FVC from baseline to week 156 with 95% CIs estimated using the Wald method. The safety endpoint-occurrence of adverse events-will be assessed in each system organ class/preferred term. ETHICS AND DISSEMINATION The study protocol and informed consent documents were approved by the Institutional Review Board at Nagasaki University Hospital (approval number 19102146) and each participating site. Written informed consent was obtained from all participants. Patient recruitment has begun. The results will be disseminated through scientific peer-reviewed publications and national and international conferences. TRIAL REGISTRATION NUMBER UMIN000038192.
Collapse
Affiliation(s)
- Noriho Sakamoto
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Hamada
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Okamoto
- Division of Respirology, Neurology and Rheumatology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Department of Respirology and Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Kazunori Tobino
- Department of Respiratory Medicine, Iizuka Hospital, Iizuka, Japan
| | - Hidenori Ichiyasu
- Department of Respiratory Medicine, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Hiroshi Ishii
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Chikushino, Japan
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Shimpei Morimoto
- Innovation Platform & Office for Precision Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Naoki Hosogaya
- Clinical Research Center, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
19
|
Planté-Bordeneuve T, Pilette C, Froidure A. The Epithelial-Immune Crosstalk in Pulmonary Fibrosis. Front Immunol 2021; 12:631235. [PMID: 34093523 PMCID: PMC8170303 DOI: 10.3389/fimmu.2021.631235] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Interactions between the lung epithelium and the immune system involve a tight regulation to prevent inappropriate reactions and have been connected to several pulmonary diseases. Although the distal lung epithelium and local immunity have been implicated in the pathogenesis and disease course of idiopathic pulmonary fibrosis (IPF), consequences of their abnormal interplay remain less well known. Recent data suggests a two-way process, as illustrated by the influence of epithelial-derived periplakin on the immune landscape or the effect of macrophage-derived IL-17B on epithelial cells. Additionally, damage associated molecular patterns (DAMPs), released by damaged or dying (epithelial) cells, are augmented in IPF. Next to “sterile inflammation”, pathogen-associated molecular patterns (PAMPs) are increased in IPF and have been linked with lung fibrosis, while outer membrane vesicles from bacteria are able to influence epithelial-macrophage crosstalk. Finally, the advent of high-throughput technologies such as microbiome-sequencing has allowed for the identification of a disease-specific microbial environment. In this review, we propose to discuss how the interplays between the altered distal airway and alveolar epithelium, the lung microbiome and immune cells may shape a pro-fibrotic environment. More specifically, it will highlight DAMPs-PAMPs pathways and the specificities of the IPF lung microbiome while discussing recent elements suggesting abnormal mucosal immunity in pulmonary fibrosis.
Collapse
Affiliation(s)
- Thomas Planté-Bordeneuve
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Charles Pilette
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Antoine Froidure
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| |
Collapse
|
20
|
Gomes PS, Soares MR, Marchenta MFML, Meirelles GDSP, Ferreira RG, Botelho AB, Martins RB, Pereira CADC. Carbohydrate antigen 15-3 as a marker of disease severity in patients with chronic hypersensitivity pneumonitis. ACTA ACUST UNITED AC 2021; 47:e20200589. [PMID: 33681879 PMCID: PMC8332669 DOI: 10.36416/1806-3756/e20200589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/04/2021] [Indexed: 12/02/2022]
Abstract
Objective: Biomarkers associated with mucin 1, such as Krebs von den Lungen-6 and carbohydrate antigen (CA) 15-3, are increased in various interstitial lung diseases. Our aim was to determine whether CA 15-3 could be considered a biomarker of disease severity in patients with chronic hypersensitivity pneumonitis (cHP). Methods: This was a prospective observational study involving adult patients with cHP. Serum levels of CA 15-3 were measured and were correlated with variables related to disease severity and extension. HRCT scans were quantitatively analyzed using a computational platform and an image analysis tool (Computer Aided Lung Informatics for Pathology Evaluation and Rating). CA 15-3 levels were normalized by logarithmic transformation. Results: The sample comprised 41 patients. The mean age of the patients was 60.1 ± 11.6 years. The mean FVC in % of predicted was 70.3% ± 17.3%, and the median of the serum level of CA 15-3 was 48.1 U/mL. CA 15-3 levels inversely correlated with FVC in % of predicted (r = −0,30; p = 0,05), DLCO in % of predicted (r = −0,54; p < 0,01), and SpO2 at the end of a 4-min step test (r = −0,59; p < 0,01), but they directly correlated with total quantitative HRCT scores (r = 0,47; p = 0,004), especially regarding ground-glass opacities (r = 0.58; p < 0,001). Conclusions: CA 15-3 is likely to be a biomarker of disease severity of patients with cHP, particularly regarding gas exchange abnormalities.
Collapse
Affiliation(s)
- Paula Silva Gomes
- . Disciplina de Pneumologia, Universidade Federal de São Paulo - UNIFESP - São Paulo (SP) Brasil
| | - Maria Raquel Soares
- . Disciplina de Pneumologia, Universidade Federal de São Paulo - UNIFESP - São Paulo (SP) Brasil
| | | | | | - Rimarcs Gomes Ferreira
- . Disciplina de Patologia, Universidade Federal de São Paulo - UNIFESP - São Paulo (SP) Brasil
| | - André Bezerra Botelho
- . Disciplina de Pneumologia, Universidade Federal de São Paulo - UNIFESP - São Paulo (SP) Brasil
| | | | | |
Collapse
|
21
|
Tanaka N, Nishimura K, Waki D, Kadoba K, Murabe H, Yokota T. Annual variation rate of KL-6 for predicting acute exacerbation in patients with rheumatoid arthritis-associated interstitial lung disease. Mod Rheumatol 2021; 31:1100-1106. [PMID: 33496209 DOI: 10.1080/14397595.2021.1879346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVES This study evaluated the prognostic factors for acute exacerbation (AE), including sequential changes in Krebs von den Lungen-6 (KL-6) levels, in rheumatoid arthritis-associated interstitial lung disease (RA-ILD) patients. METHODS This was a retrospective observational study. We reviewed 125 patients diagnosed with RA-ILD between 2010 and 2019. We defined ΔKL-6 as the annual variation rate of KL-6 one visit before AE onset (or the last visit). The Cox regression analysis was used for evaluating significant variables associated with AE. We analysed the overall survival and respiratory-related death-free survival. RESULTS Thirty-three patients (26.4%) developed AE during the observation period. The univariate analysis revealed that KL-6 levels at RA-ILD diagnosis [hazard ratio (HR), 1.11; 95% confidence interval (CI), 1.05-1.15; p < .01) and ΔKL-6 (HR: 3.69; 95% CI: -1.36 to 7.96; p = .01] were significantly associated with AE. ΔKL-6 was an independent prognostic factor for AE in the multivariate analysis (HR: 3.37; 95% CI: -1.16 to 8.87; p = .03). Patients with AE had a significantly higher overall mortality rate (p = .02) and respiratory-related mortality rate (p < .01) than those without AE. CONCLUSION ΔKL-6 can be a prognostic marker for detecting AE in RA-ILD patients.
Collapse
Affiliation(s)
- Nozomi Tanaka
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Keisuke Nishimura
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Daisuke Waki
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Keiichiro Kadoba
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Hiroyuki Murabe
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Toshihiko Yokota
- Department of Endocrinology and Rheumatology, Kurashiki Central Hospital, Kurashiki, Japan
| |
Collapse
|
22
|
Gono T, Masui K, Nishina N, Kawaguchi Y, Kawakami A, Ikeda K, Kirino Y, Sugiyama Y, Tanino Y, Nunokawa T, Kaneko Y, Sato S, Asakawa K, Ukichi T, Kaieda S, Naniwa T, Okano Y, Kuwana M. Risk Prediction Modeling Based on a Combination of Initial Serum Biomarker Levels in Polymyositis/Dermatomyositis-Associated Interstitial Lung Disease. Arthritis Rheumatol 2021; 73:677-686. [PMID: 33118321 DOI: 10.1002/art.41566] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 10/22/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To establish predictive models for mortality in patients with polymyositis/dermatomyositis-associated interstitial lung disease (PM/DM-ILD) using a combination of initial serum biomarker levels. METHODS The Multicenter Retrospective Cohort of Japanese Patients with Myositis-Associated ILD (JAMI) database of 497 incident cases of PM/DM-ILD was used as a derivation cohort, and 111 cases were additionally collected as a validation cohort. Risk factors predictive of all-cause mortality were identified by univariate and multivariable Cox regression analyses using candidate serum biomarkers as explanatory variables. The predictive models for mortality were generated in patients with and those without anti-melanoma differentiation-associated gene 5 (MDA-5) antibody, using a combination of risk factors. Cumulative survival rates were assessed using Kaplan-Meier analysis, and were compared between subgroups using the Breslow test. RESULTS In the derivation cohort, C-reactive protein (CRP) and Krebs von den Lungen 6 (KL-6) levels were identified as independent risk factors for mortality in both anti-MDA-5-positive and anti-MDA-5-negative patients. We then developed a prediction model based on anti-MDA-5 antibody status, CRP level, and KL-6 level, termed the "MCK model," to identify patients at low (<15%), moderate (15-50%), or high (≥50%) risk of mortality, based on the number of risk factors. The MCK model successfully differentiated cumulative survival rates in anti-MDA-5-positive patients (P < 0.01 for low versus moderate risk and P = 0.03 for moderate versus high risk) and in anti-MDA-5-negative patients (P < 0.001 for low versus moderate risk). The utility of the MCK model was replicated in the validation cohort. CONCLUSION Our findings indicate that an evidence-based risk prediction model using CRP and KL-6 levels combined with anti-MDA-5 antibody status might be useful for predicting prognosis in patients with PM/DM-ILD.
Collapse
Affiliation(s)
- Takahisa Gono
- Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Kenichi Masui
- National Defense Medical College School of Medicine, Saitama, Japan, and, Show University Hospital, Tokyo, Japan
| | | | | | - Atsushi Kawakami
- Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kei Ikeda
- Chiba University Hospital, Chiba, Japan
| | - Yohei Kirino
- Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Yoshinori Tanino
- Fukushima Medical University School of Medicine, Fukushima, Japan
| | | | - Yuko Kaneko
- Keio University School of Medicine, Tokyo, Japan
| | - Shinji Sato
- Tokai University School of Medicine, Kanagawa, Japan
| | | | - Taro Ukichi
- The Jikei University School of Medicine, Tokyo, Japan
| | | | - Taio Naniwa
- Nagoya City University School of Medicine, Aichi, Japan
| | | | - Masataka Kuwana
- Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | | |
Collapse
|
23
|
Mori Y, Kondoh Y. What parameters can be used to identify early idiopathic pulmonary fibrosis? Respir Investig 2021; 59:53-65. [PMID: 33277230 DOI: 10.1016/j.resinv.2020.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 06/12/2023]
Abstract
Elucidating the disease process of early idiopathic pulmonary fibrosis (IPF) will help clinicians in addressing the contentious issues of when and in which patients, therapeutic intervention should be initiated. Here, we discuss several possible parameters for diagnosing early IPF and their clinical impacts. Physiologically, early IPF can be considered as IPF with normal or mild impairment in pulmonary function. Radiologically, early IPF can be considered as IPF with a small extent and/or early features of fibrosis. Symptomatically, early IPF can be considered as asymptomatic or less symptomatic IPF. IPF at Gender-Age-Physiology index stage I can be considered early IPF. Interstitial lung abnormalities are defined as parenchymal abnormalities in more than 5% of the lung in patients with no prior history of interstitial lung disease, and in some cases, this seems to be equivalent to early IPF. Previous clinical trials showed the effect of antifibrotic therapies in early IPF, but the effects of therapy are uncertain in early IPF outside of clinical trials, such as in cases of IPF with normal pulmonary function, IPF without honeycombing or traction bronchiectasis, and asymptomatic IPF. Moreover, little has been reported on disease progression in such conditions. Because the conceptual framework of early IPF may vary depending on its definition, not only is a diagnosis of early IPF important but prediction of disease progression is also crucial. Further investigations are needed to identify biomarkers that can detect patients who may experience greater degrees of disease progression and require treatment even with those forms of early IPF.
Collapse
Affiliation(s)
- Yuta Mori
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi, 489-8642, Japan; Department of Respiratory Medicine, Ogaki Municipal Hospital, Ogaki, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi, 489-8642, Japan.
| |
Collapse
|
24
|
Ikeda K, Chiba H, Nishikiori H, Azuma A, Kondoh Y, Ogura T, Taguchi Y, Ebina M, Sakaguchi H, Miyazawa S, Suga M, Sugiyama Y, Nukiwa T, Kudoh S, Takahashi H. Serum surfactant protein D as a predictive biomarker for the efficacy of pirfenidone in patients with idiopathic pulmonary fibrosis: a post-hoc analysis of the phase 3 trial in Japan. Respir Res 2020; 21:316. [PMID: 33256760 PMCID: PMC7706186 DOI: 10.1186/s12931-020-01582-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive, fatal disorder with a variable disease course. The recent advancement of antifibrotic therapy has increased the need for reliable and specific biomarkers. This study aimed to assess alveolar epithelial biomarkers as predictors for the efficacy of the antifibrotic drug pirfenidone. Methods We conducted a post-hoc analysis of the prospective, multicenter, randomized, placebo-controlled, phase 3 trial of pirfenidone in Japan (total, n = 267; pirfenidone, n = 163; placebo, n = 104). Logistic regression analysis was performed to extract parameters that predicted disease progression, defined by a ≥ 10% relative decline in vital capacity (VC) from baseline and/or death, at week 52. For assessment of serum surfactant protein (SP)-D, SP-A and Krebs von den Lungen (KL)-6, all patients were dichotomized by the median concentration of each biomarker at baseline to the high and low biomarker subgroups. Associations of these concentrations were examined with changes in VC at each time point from baseline up to week 52, along with progression-free survival (PFS). Additionally, the effect of pirfenidone treatment on serial longitudinal concentrations of these biomarkers were evaluated. Results In the multivariate logistic regression analysis, body mass index (BMI), %VC and SP-D in the pirfenidone group, and BMI and %VC in the placebo group were indicated as predictors of disease progression. Pirfenidone treatment reduced the decline in VC with statistical significance in the low SP-D and low SP-A subgroups over most of the treatment period, and also prolonged PFS in the low SP-D and low KL-6 subgroups. Furthermore, SP-D levels over time course were reduced in the pirfenidone group from as early as week 8 until the 52-week treatment period compared with the placebo group. Conclusions Serum SP-D was the most consistent biomarker for the efficacy of pirfenidone in the cohort trial of IPF. Serial measurements of SP-D might have a potential for application as a pharmacodynamic biomarker. Trial registration The clinical trial was registered with the Japan Pharmaceutical Information Center (JAPIC) on September 13, 2005 (registration No. JapicCTI-050121; http://Clinicaltrials.jp)
Collapse
Affiliation(s)
- Kimiyuki Ikeda
- Department of Respiratory Medicine and Allergology, School of Medicine, Sapporo Medical University, South 1, West 16, Sapporo, 060-8543, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, School of Medicine, Sapporo Medical University, South 1, West 16, Sapporo, 060-8543, Japan.
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, School of Medicine, Sapporo Medical University, South 1, West 16, Sapporo, 060-8543, Japan
| | | | | | - Takashi Ogura
- Kanagawa Cardiovascular and Respiratory Center, Yokohama, Japan
| | | | - Masahito Ebina
- Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | | | | | | | | | | | - Shoji Kudoh
- Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, School of Medicine, Sapporo Medical University, South 1, West 16, Sapporo, 060-8543, Japan
| | | |
Collapse
|
25
|
Kameda M, Otsuka M, Chiba H, Kuronuma K, Hasegawa T, Takahashi H, Takahashi H. CXCL9, CXCL10, and CXCL11; biomarkers of pulmonary inflammation associated with autoimmunity in patients with collagen vascular diseases-associated interstitial lung disease and interstitial pneumonia with autoimmune features. PLoS One 2020; 15:e0241719. [PMID: 33137121 PMCID: PMC7605704 DOI: 10.1371/journal.pone.0241719] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/20/2020] [Indexed: 11/19/2022] Open
Abstract
Introduction Interstitial lung disease (ILD) is a heterogeneous group of diseases characterized by varying degrees of lung inflammation and/or fibrosis. We investigated biomarkers to infer whether patients with collagen vascular diseases associated ILD (CVD–ILD) and interstitial pneumonia with autoimmune features (IPAF) benefit from immunosuppressive therapy. Materials and methods We retrospectively investigated patients with CVD–ILD, IPAF, and idiopathic pulmonary fibrosis (IPF) between June 2013 and May 2017 at our department. First, we assessed differences in serum and bronchoalveolar lavage fluid (BALF) levels of cytokines between groups. Second, we assessed the associations of patient’s clinical variables with serum and BALF levels of those cytokines that were different between groups. Finally, we assessed the associations of diagnosis and response to immunosuppressive therapy with serum levels of those cytokines that were different between groups. Results We included 102 patients (51 with IPF, 35 with IPAF, and 16 with CVD–ILD). Serum and BALF levels of CXCL9, CXCL10, and CXCL11 were significantly elevated in patients with IPAF or CVD–ILD compared with those in patients with IPF. BALF levels of CXCL9 and CXCL10 were correlated with the percentages of lymphocytes and macrophages in BALF. Serum levels of CXCL9 and CXCL10 were correlated with BALF levels. Serum levels of CXCL9, CXCL10, and CXCL11 were correlated C-reactive protein, percent predicted forced vital capacity, alveolar-arterial oxygen difference, and the percentages of lymphocytes and macrophages in BALF. Serum levels of CXCL9, CXCL10, and CXCL11 showed moderate accuracy to distinguish patients with CVD–ILD from those with IPAF and IPF. Pre-treatment serum levels of CXCL9 and CXCL11 showed strong positive correlations with the annual forced vital capacity changes in patients with IPAF and CVD–ILD treated with immunosuppressive drugs. Conclusions Serum CXCL9, CXCL10, and CXCL11 are potential biomarkers for autoimmune inflammation and predictors of the immunosuppressive therapy responses in ILD with background autoimmunity.
Collapse
Affiliation(s)
- Masami Kameda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine, Sapporo-Kosei General Hospital, Sapporo, Hokkaido, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
- * E-mail:
| | | | - Hiroki Takahashi
- Department of Rheumatology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
26
|
Pro-fibrotic Factors as Potential Biomarkers of Anti-fibrotic Drug Therapy in Patients With Idiopathic Pulmonary Fibrosis. Arch Bronconeumol 2020; 57:231-233. [PMID: 32981796 DOI: 10.1016/j.arbres.2020.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 11/24/2022]
|
27
|
King SD, Chen SY. Recent progress on surfactant protein A: cellular function in lung and kidney disease development. Am J Physiol Cell Physiol 2020; 319:C316-C320. [PMID: 32639871 DOI: 10.1152/ajpcell.00195.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Pulmonary surfactant is a heterogeneous active surface complex made up of lipids and proteins. The major glycoprotein in surfactant is surfactant protein A (SP-A), which is released into the alveolar lumen from cytoplasmic lamellar bodies in type II alveolar epithelial cells. SP-A is involved in phospholipid absorption. SP-A together with other surfactant proteins and phospholipids prevent alveolar collapse during respiration by decreasing the surface tension of the air-liquid interface. Additionally, SP-A interacts with pathogens to prevent their propagation and regulate host immune responses. Studies in human and animal models have shown that deficiencies or mutations in surfactant components result in various lung or kidney pathologies, suggesting a role for SP-A in the development of lung and kidney diseases. In this mini-review, we discuss the current understanding of SP-A functions, recent findings of its dysfunction in specific lung and kidney pathologies, and how SP-A has been used as a biomarker to detect the outcome of lung diseases.
Collapse
Affiliation(s)
- Skylar D King
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri
| | - Shi-You Chen
- Department of Surgery, University of Missouri School of Medicine, Columbia, Missouri.,Department of Molecular Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| |
Collapse
|
28
|
Kaieda S, Gono T, Masui K, Nishina N, Sato S, Kuwana M. Evaluation of usefulness in surfactant protein D as a predictor of mortality in myositis-associated interstitial lung disease. PLoS One 2020; 15:e0234523. [PMID: 32525903 PMCID: PMC7289364 DOI: 10.1371/journal.pone.0234523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/26/2020] [Indexed: 11/30/2022] Open
Abstract
Objective Surfactant protein D (SP-D) is considered a serum biomarker of various forms of interstitial lung disease (ILD). In this study, we examined the utility of SP-D as a predictive biomarker for mortality in patients with ILD associated with polymyositis/dermatomyositis (PM/DM) using large-scale multicentre cohort data. Methods We enrolled 381 patients with incident PM/DM-associated ILD in a multicentre retrospective cohort based on the availability of serum SP-D at the baseline. Demographic and clinical characteristics as well as the presence of autoantibodies to melanoma differentiation-associated gene 5 (MDA5) and aminoacyl tRNA synthetase were measured at the time of diagnosis, and follow-up survival data were collected prospectively. Results Seventy-eight patients died during the median observation period of 18 months, and the majority of patients died of ILD. The SP-D levels at baseline were significantly lower (P = 0.02) in a non-survivor subset than in a survivor subset among the entire enrolled patients. However, the SP-D levels were higher in the non-survivor subset than in the survivor subset based on the stratification by anti-MDA5-positive, anti-ARS-positive and, double-negativity, although there was an only statistically significant difference (P = 0.01) in the double-negative group. Surprisingly, the SP-D levels were within the upper limit of normal, 110 ng/mL, in 54 (87%) of 62 anti-MDA5-positive patients who died. In the double-negative group, the mortality rates were significantly higher (P = 0.002) in a subset with SP-D ≥127.6 ng/mL, the cut-off value for mortality calculated by the receiver operating characteristic curve, than the other subset. All of patients with SP-D <127.6 ng/mL survived. Conclusion Serum SP-D levels behave differently among patients with stratified by anti-MDA5 antibody, anti-ARS antibody and both negativity in PM/DM-associated ILD. Its use in clinical practice should be applied with caution on the basis of the presence or absence of anti-MDA5 antibody or anti-ARS antibody.
Collapse
Affiliation(s)
- Shinjiro Kaieda
- Department of Medicine, Division of Respirology, Neurology, and Rheumatology, Kurume University School of Medicine, Fukuoka, Japan
| | - Takahisa Gono
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
- * E-mail:
| | - Kenichi Masui
- Department of Anaesthesiology, Showa University School of Medicine, Tokyo, Japan
| | - Naoshi Nishina
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shinji Sato
- Division of Rheumatology, Department of Internal Medicine, Tokai University School of Medicine, Tokyo, Kanagawa, Japan
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | | |
Collapse
|
29
|
Landi C, Bergantini L, Cameli P, d'Alessandro M, Carleo A, Shaba E, Rottoli P, Bini L, Bargagli E. Idiopathic Pulmonary Fibrosis Serum proteomic analysis before and after nintedanib therapy. Sci Rep 2020; 10:9378. [PMID: 32523095 PMCID: PMC7287088 DOI: 10.1038/s41598-020-66296-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/16/2020] [Indexed: 11/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal progressive disease with a median survival of 2–5 years. Nintedanib is a small tyrosine kinase inhibitor that reduces IPF progression, significantly slowing the annual decline in Forced Vital Capacity (FVC). Very little data is available on the molecular mechanisms of this treatment in IPF, despite a growing interest in the definition of IPF pathogenesis and target therapy. A functional proteomic approach was applied to the analysis of serum samples from IPF patients in order to highlight differential proteins potentially indicative of drug-induced molecular pathways modifications and response to therapy. Twelve serum samples were collected from six IPF patients in care at Siena Regional Referral Center for Interstitial Lung Diseases (ILDs) and treated with nintedanib for one year. Serum samples were analyzed at baseline (T0 before starting therapy) and after one year of treatment (T1) and underwent differential proteomic and bioinformatic analysis. Proteomic analysis revealed 13 protein species that were significantly increased after one year of treatment. When the targets of nintedanib (VEGFR, FGFR and PDGFR) were added, enrichment analysis extracted molecular pathways and process networks involved in cell differentiation (haptoglobin and albumin), coagulation (antithrombin III), epithelial mesenchymal transition, cell proliferation and transmigration. PI3K and MAPK induced up-regulation of apolipoprotein C3. Proteomic study found 13 protein species up-regulated in IPF patients after one year of nintedanib treatment. Haptoglobin, a central hub of our analysis was validated by 2D-WB and ELISA as theranostic marker in a more numerous populations of patients.
Collapse
Affiliation(s)
- Claudia Landi
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy. .,Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy.
| | - Laura Bergantini
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Paolo Cameli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Miriana d'Alessandro
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Alfonso Carleo
- Department of Pneumology, Medical School Hannover (MHH), Hannover, Germany
| | - Enxhi Shaba
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy
| | - Paola Rottoli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| | - Luca Bini
- Functional Proteomics Lab, Dept. Life Sciences, University of Siena, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplant Unit, Dept. Internal and Specialist Medicine, AOUS, Siena, Italy
| |
Collapse
|
30
|
Diagnostic and Prognostic Biomarkers for Chronic Fibrosing Interstitial Lung Diseases With a Progressive Phenotype. Chest 2020; 158:646-659. [PMID: 32268131 DOI: 10.1016/j.chest.2020.03.037] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/05/2020] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Biomarkers have the potential to become central to the clinical evaluation and monitoring of patients with chronic fibrosing interstitial lung diseases (ILDs) with a progressive phenotype. Here we summarize the current understanding of putative serum, BAL fluid, and genetic biomarkers in this setting, according to their hypothesized pathobiologic mechanisms: evidence of epithelial cell dysfunction (eg, Krebs von den Lungen-6 antigen), fibroblast proliferation and extracellular matrix production or turnover (eg, matrix metalloproteinase-1), or immune dysregulation (eg, CC chemokine ligand 18). While most of the available data come from idiopathic pulmonary fibrosis (IPF), the prototypic progressive fibrosing ILD, data are available in the broader patient population of chronic fibrosing ILDs. A number of these biomarkers show promise, however, none have been validated. In this review article, we assess both the status of proposed biomarkers for chronic fibrosing lung diseases with a progressive phenotype in predicting disease risk or predisposition, diagnosis, prognosis, and treatment response and provide a direct comparison between IPF and other chronic fibrotic ILDs. We also reflect on the current clinical usefulness and future direction of research for biomarkers in the setting of chronic fibrosing ILDs with a progressive phenotype.
Collapse
|
31
|
Yoshikawa T, Otsuka M, Chiba H, Ikeda K, Mori Y, Umeda Y, Nishikiori H, Kuronuma K, Takahashi H. Surfactant protein A as a biomarker of outcomes of anti-fibrotic drug therapy in patients with idiopathic pulmonary fibrosis. BMC Pulm Med 2020; 20:27. [PMID: 32005219 PMCID: PMC6995128 DOI: 10.1186/s12890-020-1060-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/23/2020] [Indexed: 01/19/2023] Open
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is a progressive and fibrosing lung disease with poor prognosis. Pirfenidone and nintedanib are anti-fibrotic drugs used for patients with IPF. These drugs reduce the rate of decline in forced vital capacity (FVC). Serum surfactant protein (SP)-A, SP-D, and Krebs von den Lungen-6 (KL-6) are monitoring and prognostic biomarkers in patients with IPF; however, their relationship with the therapeutic outcomes of anti-fibrotic drugs has not been investigated. We aim to clarify whether serum SP-A, SP-D, and KL-6 reflect therapeutic outcomes of pirfenidone and nintedanib administration in patients with IPF. Methods We retrospectively investigated patients with IPF who were initiated on pirfenidone or nintedanib administration between January 2014 and June 2018 at our hospital. Changes in clinical parameters and serum SP-A, SP-D, and KL-6 levels were evaluated. Patients with ≥10% decline in FVC or ≥ 15% decline in diffusing capacity of the lung for carbon monoxide (DLco) from baseline to 6 months were classified as progression group, while the other patients were classified as stable group. Results Forty-nine patients were included (pirfenidone, 23; nintedanib, 26). Stable group comprised 32 patients, while progression group comprised 17 patients. In the stable group, changes in SP-A and KL-6 from baseline to 3 and 6 months significantly decreased compared with the progression group (SP-A: 3 months − 6.0% vs 16.7%, 6 months − 10.2% vs 20.2%, KL-6: 3 months − 9.2% vs 6.7%, 6 months − 15.0% vs 12.1%, p < 0.05). Changes in SP-A and SP-D levels showed significant negative correlations with the change in %FVC (r = − 0.46 and r = − 0.39, p < 0.01, respectively) and %DLco (r = − 0.67 and r = − 0.54, p < 0.01, respectively). Similar results were also seen in subgroup analysis for both pirfenidone and nintedanib groups. On logistic regression analysis, decrease in SP-A from baseline to 3 months and 6 months was found to predict the outcomes at 6 months (odds ratios: 0.89 and 0.88, respectively). Conclusions Changes in serum SP-A reflected the outcomes of anti-fibrotic drug therapy. Serum SP-A has a potential as a biomarker of therapeutic outcomes of anti-fibrotic drugs.
Collapse
Affiliation(s)
- Takumi Yoshikawa
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan.
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Kimiyuki Ikeda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yuki Mori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Yasuaki Umeda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, 1-37, South 1-West 16, Chuo-ku, Sapporo, Hokkaido, 060-8543, Japan
| |
Collapse
|
32
|
Elhai M, Avouac J, Allanore Y. Circulating lung biomarkers in idiopathic lung fibrosis and interstitial lung diseases associated with connective tissue diseases: Where do we stand? Semin Arthritis Rheum 2020; 50:480-491. [PMID: 32089354 DOI: 10.1016/j.semarthrit.2020.01.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 01/09/2020] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
Interstitial lung diseases (ILDs) are complex diseases with various courses where personalized medicine is highly expected. Biomarkers are indicators of physiological, pathological processes or of pharmacological response to therapeutic interventions. They can be used for diagnosis, risk-stratification, prediction and monitoring of treatment response. To better delineate the input and pitfalls of biomarkers in ILDs, we performed a systematic review and meta-analysis of literature in MEDLINE and Embase databases from January 1960 to February 2019. We focused on circulating biomarkers as having the highest generalizability. Overall, 70 studies were included in the review and 20 studies could be included in the meta-analysis. This review highlights that ILD associated with connective tissue diseases (CTD-ILD) and idiopathic pulmonary fibrosis (IPF) share common biomarkers, suggesting common pathophysiological pathways. KL-6 and SP-D, could diagnose lung fibrosis in both IPF and CTD-ILD, with KL-6 having the strongest value (OR: 520.95[110.07-2465.58], p<0.001 in IPF and OR:26.43[7.15-97.68], p<0.001 in CTD-ILD), followed by SPD (OR: 33.81[3.20-357.52], p = 0.003 in IPF and 13.24 [3.84-45.71] in SSc-ILD), MMP7 appeared as interesting for IPF diagnosis (p<0.001), whereas in SSc, CCL18 was associated with ILD diagnosis. Both CCL18 and KL-6 were predictive for the outcomes of ILDs, with higher predictive values for CCL18 in both IPF (OR:10.22[4.72-22.16], p<0.001 and in SSc [2.62[1.71-4.03], p<0.001). However, disease specific biomarkers are lacking and large longitudinal studies are needed before the translational use of the potential biomarkers in clinical practice. With the recent availability of new effective therapies in ILDs, further studies should assess response to treatment.
Collapse
Affiliation(s)
- Muriel Elhai
- INSERM U1016, Rheumatology A department, Cochin Hospital, Paris Descartes University, 27 rue du Faubourg Saint Jacques, 75014 Paris, France.
| | - Jérôme Avouac
- INSERM U1016, Rheumatology A department, Cochin Hospital, Paris Descartes University, 27 rue du Faubourg Saint Jacques, 75014 Paris, France.
| | - Yannick Allanore
- INSERM U1016, Rheumatology A department, Cochin Hospital, Paris Descartes University, 27 rue du Faubourg Saint Jacques, 75014 Paris, France.
| |
Collapse
|
33
|
Nakano H, Inoue S, Shibata Y, Abe K, Murano H, Yang S, Machida H, Sato K, Sato C, Nemoto T, Nishiwaki M, Kimura T, Yamauchi K, Sato M, Igarashi A, Tokairin Y, Watanabe M. E-selectin as a prognostic factor of patients hospitalized due to acute inflammatory respiratory diseases: a single institutional study. EXCLI JOURNAL 2019; 18:1062-1070. [PMID: 31839762 PMCID: PMC6909376 DOI: 10.17179/excli2019-1624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/04/2019] [Indexed: 11/10/2022]
Abstract
When examining patients with acute inflammatory respiratory diseases, it is difficult to distinguish between infectious pneumonia and interstitial pneumonia and predict patient prognosis at the beginning of treatment. In this study, we assessed whether endothelial selectin (E-selectin) predicts the outcome of patients with acute inflammatory respiratory diseases. We measured E-selectin serum levels in 101 patients who were admitted to our respiratory care unit between January 2013 and December 2013 because of acute inflammatory respiratory diseases that were eventually diagnosed as interstitial pneumonia (n = 38) and lower respiratory tract infection (n = 63). Seven of these patients (n = 101) died. The pneumonia severity score was significantly higher and the oxygen saturation of arterial blood measured by pulse oximeter (SpO2)/fraction of inspiratory oxygen (FiO2) was significantly lower in the deceased patients than in the surviving patients. There were significantly fewer peripheral lymphocytes and significantly higher E-selectin serum levels in the deceased patients than in the surviving patients. In the multiple logistic regression analysis, the E-selectin serum levels and SpO2/FiO2 ratio were independent predictive factors of prognosis. The risk of death during acute respiratory disease was determined using a receiver operating characteristic (ROC) curve analysis. The area under the curve (AUC) was 0.871 as calculated from the ES, and the cutoff value was 6453.04 pg/ml, with a sensitivity of 1.00 and a specificity of 0.72 (p = 0.0027). E-selectin may be a useful biomarker for predicting the prognosis of patients with acute inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Hiroshi Nakano
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Sumito Inoue
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yoko Shibata
- Department of Pulmonary Medicine, Fukushima Medical University, Fukushima, Japan
| | - Koya Abe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hiroaki Murano
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Sujeong Yang
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hiroyoshi Machida
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Kento Sato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Chisa Sato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Takako Nemoto
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Michiko Nishiwaki
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Tomomi Kimura
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Keiko Yamauchi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masamichi Sato
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Akira Igarashi
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yoshikane Tokairin
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masafumi Watanabe
- Department of Cardiology, Pulmonology, and Nephrology, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
34
|
Somogyi V, Chaudhuri N, Torrisi SE, Kahn N, Müller V, Kreuter M. The therapy of idiopathic pulmonary fibrosis: what is next? Eur Respir Rev 2019; 28:190021. [PMID: 31484664 PMCID: PMC9488691 DOI: 10.1183/16000617.0021-2019] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fibrosing interstitial lung disease, characterised by progressive scarring of the lung and associated with a high burden of disease and early death. The pathophysiological understanding, clinical diagnostics and therapy of IPF have significantly evolved in recent years. While the recent introduction of the two antifibrotic drugs pirfenidone and nintedanib led to a significant reduction in lung function decline, there is still no cure for IPF; thus, new therapeutic approaches are needed. Currently, several clinical phase I-III trials are focusing on novel therapeutic targets. Furthermore, new approaches in nonpharmacological treatments in palliative care, pulmonary rehabilitation, lung transplantation, management of comorbidities and acute exacerbations aim to improve symptom control and quality of life. Here we summarise new therapeutic attempts and potential future approaches to treat this devastating disease.
Collapse
Affiliation(s)
- Vivien Somogyi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Nazia Chaudhuri
- Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK
| | - Sebastiano Emanuele Torrisi
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
- Regional Referral Centre for Rare Lung Diseases, University Hospital "Policlinico", Dept of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Nicolas Kahn
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Veronika Müller
- Dept of Pulmonology, Semmelweis University, Budapest, Hungary
| | - Michael Kreuter
- Center for Interstitial and Rare Lung Diseases, Thoraxklinik, University of Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
35
|
Krauss E, Froehler M, Degen M, Mahavadi P, Dartsch RC, Korfei M, Ruppert C, Seeger W, Guenther A. Exhalative Breath Markers Do Not Offer for Diagnosis of Interstitial Lung Diseases: Data from the European IPF Registry (eurIPFreg) and Biobank. J Clin Med 2019; 8:jcm8050643. [PMID: 31075945 PMCID: PMC6572439 DOI: 10.3390/jcm8050643] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 04/26/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023] Open
Abstract
Background: New biomarkers are urgently needed to facilitate diagnosis in Interstitial Lung Diseases (ILD), thus reducing the need for invasive procedures, and to enable tailoring and monitoring of medical treatment. Methods: In this study we investigated if patients with idiopathic pulmonary fibrosis (IPF; n = 21), non-IPF ILDs (n = 57) and other lung diseases (chronic obstructive pulmonary disease (COPD) n = 24, lung cancer (LC) n = 16) as well as healthy subjects (n = 20) show relevant differences in exhaled NO (FeNO; Niox MINO), or in eicosanoid (PGE2, 8-isoprostane; enzyme-linked immunosorbent assay (ELISA)) levels as measured in exhaled breath condensates (EBC) and bronchoalveolar lavage fluids (BALF). Results: There was no significant difference in FeNO values between IPF, non-IPF ILDs and healthy subjects, although some individual patients showed highly elevated FeNO. On the basis of the FeNO signal, it was neither possible to differentiate between the kind of disease nor to detect exacerbations. In addition, there was no correlation between FeNO values and lung function. The investigation of the eicosanoids in EBCs was challenging (PGE2) or unreliable (8-isoprostane), but worked out well in BALF. A significant increase of free 8-isoprostane was observed in BALF, but not in EBCs, of patients with IPF, hypersensitivity pneumonitis (HP) and sarcoidosis, possibly indicating severity of oxidative stress. Conclusions: FeNO-measurements are not of diagnostic benefit in different ILDs including IPF. The same holds true for PGE2 and 8-isoprostane in EBC by ELISA.
Collapse
Affiliation(s)
- Ekaterina Krauss
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
| | - Maike Froehler
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
| | - Maria Degen
- Agaplesion Lung Clinic, 35753 Greifenstein, Germany.
| | - Poornima Mahavadi
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
| | - Ruth C Dartsch
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
- Agaplesion Lung Clinic, 35753 Greifenstein, Germany.
| | - Martina Korfei
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
| | - Clemens Ruppert
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
| | - Werner Seeger
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
- Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Justus-Liebig University Giessen, 35394 Giessen, Germany.
| | - Andreas Guenther
- European IPF Registry & Biobank (eurIPFreg/bank), 35394 Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35394 Giessen, Germany.
- Agaplesion Lung Clinic, 35753 Greifenstein, Germany.
- Cardio-Pulmonary Institute (CPI), EXC 2026, Project ID: 390649896, Justus-Liebig University Giessen, 35394 Giessen, Germany.
| |
Collapse
|
36
|
Kishaba T. Acute Exacerbation of Idiopathic Pulmonary Fibrosis. ACTA ACUST UNITED AC 2019; 55:medicina55030070. [PMID: 30884853 PMCID: PMC6473875 DOI: 10.3390/medicina55030070] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/18/2019] [Accepted: 03/14/2019] [Indexed: 12/02/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is the most common form of idiopathic interstitial pneumonia. Idiopathic pulmonary fibrosis is often seen in elderly men who smoke. A diagnosis of IPF is based on a combination of a detailed clinical history, specific physical examination, laboratory findings, pulmonary function tests, high-resolution computed tomography (HRCT) of the chest, and histopathology. Idiopathic pulmonary fibrosis has a heterogeneous clinical course, from an asymptomatic stable state to progressive respiratory failure or acute exacerbation (AE). Acute exacerbation of IPF has several important differential diagnoses, such as heart failure and volume overload. The International Working Group project proposed new criteria for defining AE of IPF in 2016, which divides it into triggered and idiopathic AE. On the basis of these criteria, physicians can detect AE of IPF more easily. The recent international IPF guidelines emphasized the utility of chest HRCT. In addition, two antifibrotic agents have become available. We should focus on both the management and prevention of AE. The diagnostic process, laboratory findings, typical chest imaging, management, and prognosis of AE are comprehensively reviewed in this article.
Collapse
Affiliation(s)
- Tomoo Kishaba
- Department of Respiratory Medicine, Okinawa Chubu Hospital, 904-2293 Miyazato 281, Uruma City, Okinawa, Japan.
| |
Collapse
|
37
|
Kishaba T. New era of Idiopathic Pulmonary Fibrosis. Respir Investig 2018; 56:427-429. [PMID: 30366836 DOI: 10.1016/j.resinv.2018.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Tomoo Kishaba
- Okinawa Chubu Hospital, Miyazato 281, Uruma City, Okinawa, Japan.
| |
Collapse
|