1
|
Fan J, Cong S, Zhang Y, Jiang X, Wang N, Fang L, Chen Y. Associations of pre-COPD indicators with lung function decline and their longitudinal transitions. Pulmonology 2025; 31:2486881. [PMID: 40353713 DOI: 10.1080/25310429.2025.2486881] [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: 11/05/2024] [Accepted: 03/26/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Pre-chronic obstructive pulmonary disease (COPD) indicators are associated with COPD development, but their associations with lung function decline in persons without COPD and their longitudinal transitions remain uncertain. METHODS In this prospective cohort study, 3526 subjects without COPD from the 2014-2015 national COPD surveillance in China were investigated for a second time during 2018-2020. Four potential pre-COPD indicators at baseline were chronic bronchitis, preserved ratio impaired spirometry (PRISm), low peak expiratory flow (PEF), and spirometric small airway dysfunction (sSAD). To include the incident COPD subjects in the lung function decline analysis for consistent indicators, forced expiratory volume in 1 s (FEV1) <80% predicted was used instead of PRISm. Lung function outcomes were the declines in post-bronchodilator FEV1, forced vital capacity (FVC), and FEV1/FVC. RESULTS Subjects with initial chronic bronchitis had faster declines in FEV1 and FVC compared with those without initial chronic bronchitis among smokers. Consistent chronic bronchitis was associated with faster declines in FEV1 and FVC among non-smokers. Consistent sSAD was associated with faster declines in FEV1/FVC among smokers and non-smokers. Consistent FEV1 <80% predicted and consistent low PEF were associated with a faster decline in FEV1/FVC among smokers. Incident COPD developed from sSAD in almost half of the cases. CONCLUSIONS Initial chronic bronchitis and consistent FEV1 <80% predicted, sSAD, and low PEF are associated with excessive lung function decline among smokers, while consistent chronic bronchitis and sSAD are associated with excessive lung function decline among non-smokers. Initial sSAD accounts for a larger proportion of incident COPD than initial chronic bronchitis, PRISm, and low PEF.
Collapse
Affiliation(s)
- Jing Fan
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shu Cong
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Zhang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiao Jiang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ning Wang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liwen Fang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
2
|
Hossain R, Hossain MS, Lee HJ, Lee CJ. Hederacoside C Modulates EGF-Induced MUC5AC Mucin Gene Expression by Regulating the MAPK Signaling Pathway in Human Airway Epithelial Cells. Biomol Ther (Seoul) 2025; 33:510-517. [PMID: 40275567 PMCID: PMC12059360 DOI: 10.4062/biomolther.2025.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
This study aimed to evaluate the potential of hederacoside C, an active compound isolated from Hedera helix, which has been used for managing inflammatory respiratory diseases, in attenuating epidermal growth factor (EGF)-induced airway MUC5AC mucin gene expression. Human pulmonary mucoepidermoid NCI-H292 cells were pretreated with hederacoside C for 30 min and subsequently stimulated with EGF for 24 h. The study also examined the effect of hederacoside C on the EGF-induced mitogen-activated protein kinase (MAPK) signaling pathway. The results showed that hederacoside C inhibited MUC5AC mucin mRNA expression and the production of mucous glycoproteins by suppressing the phosphorylation of the EGF receptor (EGFR), as well as the phosphorylation of MAPK/extracellular signal-regulated kinase (ERK) 1/2 (MEK1/2), p38 MAPK, ERK 1/2 (p44/42), and the nuclear expression of specificity protein-1 (Sp1). These findings suggest that hederacoside C has the potential to reduce EGF-induced mucin gene expression by inhibiting the EGFR-MAPK-Sp1 signaling pathway in NCI-H292 cells.
Collapse
Affiliation(s)
- Rajib Hossain
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Md. Solayman Hossain
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyun Jae Lee
- Smith Liberal Arts College and Department of Addiction Science, Graduate School, Sahmyook University, Seoul 01795, Republic of Korea
| | - Choong Jae Lee
- Department of Pharmacology and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
3
|
Ozkan E, Livengood SS, Ford AA, Macdonald JK, Samir S, Klevans IW, Kesimer M. Analytical validation of total mucin concentration assay using SEC MALLS dRI for diagnosing and monitoring mucoobstructive lung diseases. Sci Rep 2025; 15:15024. [PMID: 40301448 PMCID: PMC12041582 DOI: 10.1038/s41598-025-97808-4] [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: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
Mucins play a pivotal role in the pathophysiology of mucoobstructive lung diseases. Accurate quantification of total mucin concentrations in clinical sputum samples is critical for developing objective biomarkers for diagnosis, prognosis, and therapeutic monitoring. By using sputum samples and mucin standards, the analytical performance of the measurements of total mucin concentration by Size Exclusion Chromatography coupled with Multi-Angle Laser Light Scattering and Differential Refractometer [SEC-(MALLS)-dRI] method was assessed using universal validation metrics, including precision, accuracy, recovery, parallelism, specificity, linearity, and sample stability. Possible sample contamination sources, such as saliva, blood, and DNA, were also evaluated. The method demonstrated excellent precision across low, medium, and high concentrations (CV% ≤ 2.6%) and high recovery (116%). It exhibited strong linearity over a broad dynamic range (~30-15,000 µg/mL) and stability for up to 12 months at - 20 °C in naïve samples and 4 °C in 4 M GuHCl. Measurement interference was negligible, up to 20% saliva, 2% blood, and 2% DNA. This study validates the SEC-(MALLS)-dRI method as a robust, reliable approach for quantifying total mucin concentrations in clinical sputum samples. The demonstrated analytical validity establishes its use as a biomarker platform for clinical and research applications, aiding in the diagnosis and management of hypersecretory/mucoobstructive lung diseases.
Collapse
Affiliation(s)
- Esin Ozkan
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Stephanie Sue Livengood
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Amina Ahmad Ford
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Jade Kathryn Macdonald
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Sophia Samir
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Ian William Klevans
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27517-7248, USA.
| |
Collapse
|
4
|
Hartert T, Kvysgaard JN, Thaver L, Suara-Istanbouli A, Allinson JP, Zar HJ. Understanding the childhood origins of asthma and chronic obstructive pulmonary disease: Insights from birth cohorts and studies across the life-span. J Allergy Clin Immunol 2025:S0091-6749(25)00419-1. [PMID: 40252849 DOI: 10.1016/j.jaci.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Birth cohorts have identified modifiable risk factors for asthma and respiratory health in children and adults, demonstrating the important role and pathways through which early-life events influence not only child outcomes but also adult health, disease, and mortality. This focused literature update from 2021 to 2024 summarizes birth cohort studies across the life-span that contribute to our understanding of risk factors for and the childhood origins of asthma and chronic obstructive pulmonary disease that may inform prevention efforts. We conclude that there are critical periods of developmental plasticity and susceptibility during which early-life events and exposures likely have the greatest impact on the development of asthma and chronic obstructive lung disease phenotypes, and that there are important prenatal and early childhood exposures, which, if modified, might be candidates for improving respiratory health across the life-span. Birth cohorts have been and will continue to be critical to advancing our understanding of lung health and disease across the life-span, including asthma and chronic obstructive pulmonary disease. As child mortality declines and the human population ages, data from birth cohort studies are needed to inform strategies for optimizing healthy longevity, including the investment in understanding the lifelong consequences of adverse prenatal and early childhood exposures.
Collapse
Affiliation(s)
- Tina Hartert
- Department of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tenn.
| | - Julie Nyholm Kvysgaard
- Department of Pediatrics, Copenhagen Prospective Studies on Asthma in Childhood, Herlev, and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Linesri Thaver
- Department of Pediatrics & Child Health and the SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Aisha Suara-Istanbouli
- Department of Medicine and Pediatrics, Vanderbilt University Medical Center, Nashville, Tenn
| | | | - Heather J Zar
- Department of Pediatrics & Child Health and the SA-MRC Unit on Child & Adolescent Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
5
|
Rückert-Eheberg IM, Steger A, Müller A, Linkohr B, Barthel P, Maier M, Allescher J, Sinner MF, Rizas KD, Rathmann W, Laugwitz KL, Kääb S, Peters A, Schmidt G. Respiratory rate and its associations with disease and lifestyle factors in the general population - results from the KORA-FF4 study. PLoS One 2025; 20:e0318502. [PMID: 40067853 PMCID: PMC11896064 DOI: 10.1371/journal.pone.0318502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/17/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVE The aim of the study was to derive median age- and sex-specific respiratory rates in a population-based sample of adults and to identify disease and lifestyle factors associated with elevated respiratory rates. METHODS In the population-based KORA FF4 study conducted in Augsburg, Germany, 5-minute 12-lead resting electrocardiograms (ECGpro-system, AMEDTEC) were recorded in 2,224 participants from 39 to 88 years. Respiratory rate was derived from these electrocardiograms. Sex- and age-specific medians, IQRs, and percentiles were calculated. Associations of sociodemographic, disease, and lifestyle variables with elevated resting respiratory rate were assessed by univariable and multivariable logistic regression analyses. RESULTS Respiratory rate decreased slightly from youngest to middle-aged women and men and increased in old age. Overall, median (IQR) was 15.80 (3.16) breaths per minute (brpm). Five percent of the participants had values lower than 12.06 brpm, and five percent had values above 20.06 brpm (95th percentile). Elevated respiratory rates of ≥ 18.6 brpm were found in 13.8% (n = 308). In an adjusted logistic regression model, age, abdominal obesity, diabetes, COPD, smoking, and low education were significantly associated with elevated respiratory rate. Stratified analyses showed that education appeared to be more relevant in women, while the effect of diabetes was more pronounced in men. CONCLUSIONS High respiratory rate may be an indicator of impaired health in the general population, especially regarding pulmonary and metabolic characteristics, and unfavorable lifestyle and living conditions. Individuals with an increased respiratory rate should therefore be examined and followed up more closely to recognize diseases and adverse progressions at an early stage and to possibly prevent them.
Collapse
Affiliation(s)
- Ina-Maria Rückert-Eheberg
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße, Munich-Neuherberg, Germany
| | - Alexander Steger
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
| | - Alexander Müller
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
| | - Birgit Linkohr
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße, Munich-Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
| | - Petra Barthel
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
| | - Melanie Maier
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
| | - Julia Allescher
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
| | - Moritz F. Sinner
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
- Department of Medicine I, LMU University Hospital LMU Munich, Munich, Germany,
| | - Konstantinos D. Rizas
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
- Department of Medicine I, LMU University Hospital LMU Munich, Munich, Germany,
| | - Wolfgang Rathmann
- Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, Germany,
- German Center for Diabetes Research, München-Neuherberg, Germany,
| | - Karl-Ludwig Laugwitz
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
- Department of Medicine I, LMU University Hospital LMU Munich, Munich, Germany,
| | - Stefan Kääb
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
- Department of Medicine I, LMU University Hospital LMU Munich, Munich, Germany,
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstraße, Munich-Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany,
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Georg Schmidt
- Department of Internal Medicine I, TUM School of Medicine and Health, Technical University of Munich, University Hospital, Munich, Germany,
- Department of Medicine I, LMU University Hospital LMU Munich, Munich, Germany,
| |
Collapse
|
6
|
Agustí A, Aliberti S, Blasi F, Miravitlles M, Papi A. Occluding mucous airway plugs in patients with obstructive lung diseases: a new treatable trait? ERJ Open Res 2025; 11:00793-2024. [PMID: 39872389 PMCID: PMC11770761 DOI: 10.1183/23120541.00793-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/08/2024] [Indexed: 01/30/2025] Open
Abstract
Identifying mucous plugs by chest CT should be considered carefully because it is potentially a treatable trait https://bit.ly/4gyJHFW.
Collapse
Affiliation(s)
- Alvar Agustí
- Catedra Salud Respiratoria, University of Barcelona, Barcelona, Spain
- Respiratory Institute, Clinic Barcelona, Barcelona, Spain
- FCRB-IDIBAPS Barcelona, Barcelona, Spain
- CIBER Enfermedades Respiratorias, Barcelona, Spain
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Rozzano, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
- Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marc Miravitlles
- Pneumology Department Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Alberto Papi
- Respiratory Medicine, Department of Translation Medicine, University of Ferrara, Ferrara, Italy
- CardioThoracic Clinic, Ferrara University Hospital, Ferrara, Italy
| |
Collapse
|
7
|
Hossain R, Lee HJ, Baek CH, Hwang SC, Lee CJ. Emodin Inhibited MUC5AC Mucin Gene Expression via Affecting EGFR-MAPK-Sp1 Signaling Pathway in Human Airway Epithelial Cells. Biomol Ther (Seoul) 2024; 32:736-743. [PMID: 39428383 PMCID: PMC11535290 DOI: 10.4062/biomolther.2024.160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/13/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
The aim of this study was to evaluate emodin, a natural trihydroxyanthraquinone compound found in the roots and barks of several plants including rhubarb and buckthorn, might attenuate epidermal growth factor (EGF)-induced airway MUC5AC mucin gene expression. The human pulmonary mucoepidermoid NCI-H292 cells were pretreated with for 30 min and then stimulated with EGF for the following 24 h. The effect of emodin on EGF-induced mitogen-activated protein kinase (MAPK) signaling pathway was examined. As a result, emodin blocked the expression of MUC5AC mucin mRNA and production of mucous glycoprotein via suppressing the phosphorylation of EGF receptor (EGFR), phosphorylation of mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) 1 and 2 (MEK1/2), phosphorylation of p38 MAPK, phosphorylation of ERK 1/2 (p44/42), and the nuclear expression of specificity protein-1 (Sp1). These findings imply that emodin has a potential to mitigate EGF-stimulated mucin gene expression by inhibiting the EGFR-MAPK-Sp1 signaling pathway, in NCI-H292 cells.
Collapse
Affiliation(s)
- Rajib Hossain
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Hyun Jae Lee
- Smith Liberal Arts College and Department of Addiction Science, Graduate School, Sahmyook University, Seoul 01795, Republic of Korea
| | - Chang-Heon Baek
- Department of Orthopaedic Surgery and Institute of Health Sciences, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Sun-Chul Hwang
- Department of Orthopaedic Surgery and Institute of Health Sciences, Gyeongsang National University College of Medicine and Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Choong Jae Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Brain Korea 21 FOUR Project for Medical Science, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
8
|
Jo YS, Rhee CK, Kim SH, Lee H, Choi JY. Spirometric Transition of at Risk Individuals and Risks for Progression to Chronic Obstructive Pulmonary Disease in General Population. Arch Bronconeumol 2024; 60:634-642. [PMID: 38902193 DOI: 10.1016/j.arbres.2024.05.033] [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: 02/13/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a dynamic disease with a high socioeconomic burden. Using data collected prospectively from the general population, we examined factors related to the transition of at-risk individuals to COPD. METHODS We used the Korean Genome Epidemiology Study (KoGES) database, defining pre-COPD based on respiratory symptoms and radiological abnormalities suggestive of COPD; the preserved ratio impaired spirometry (PRISm) was defined as a forced expiratory volume in 1s (FEV1)/forced vital capacity ratio≥70% and FEV1<80%, as predicted by spirometry. We determined group differences in the rate of lung function decline, risk of future airflow obstruction (AFO). RESULTS The study included 4762 individuals, and longitudinal analysis revealed distinct trends in pulmonary function indicators. Compared to the normal group, the pre-COPD group showed a more rapid decline in lung function, while the PRISm group showed a slower decline. In the pre-COPD and PRISm groups, 4.4% and 3.5%, and 13.6% and 10.8%, respectively, of patients had progressed to COPD at the first and second visits. Pre-COPD and PRISm contributed to an earlier time to first AFO, but consideration of comorbid cardiovascular disease weakened this relationship in the PRISm group. Multivariate logistic regression showed that pre-COPD and PRISm are significant risk factors for future development of COPD (OR 1.80, p<0.001; OR 4.26, p<0.001, respectively). CONCLUSION Pre-COPD and PRISm patients showed different trends in lung function changes over time and both were significant risk factors for future development of COPD.
Collapse
Affiliation(s)
- Yong Suk Jo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Hyuk Kim
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Dongguk University Gyeongju Hospital, Dongguk University College of Medicine, Gyeongju, Republic of Korea
| | - Hyun Lee
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Curtis JL, Bateman LA, Murray S, Couper DJ, Labaki WW, Freeman CM, Arnold KB, Christenson SA, Alexis NE, Kesimer M, Boucher RC, Kaner RJ, Barjaktarevic I, Cooper CB, Hoffman EA, Barr RG, Bleecker ER, Bowler RP, Comellas A, Dransfield MT, Freedman MB, Hansel NN, Krishnan JA, Marchetti N, Meyers DA, Ohar J, O'Neal WK, Ortega VE, Paine III R, Peters SP, Smith BM, Wedzicha JA, Wells JM, Woodruff PG, Han MK, Martinez FJ, for the SOURCE Investigators. Design of the SPIROMICS Study of Early COPD Progression: SOURCE Study. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2024; 11:444-459. [PMID: 39159077 PMCID: PMC11548966 DOI: 10.15326/jcopdf.2023.0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 08/21/2024]
Abstract
Background The biological mechanisms leading some tobacco-exposed individuals to develop early-stage chronic obstructive pulmonary disease (COPD) are poorly understood. This knowledge gap hampers development of disease-modifying agents for this prevalent condition. Objectives Accordingly, with National Heart, Lung and Blood Institute support, we initiated the SubPopulations and InteRmediate Outcome Measures In COPD Study (SPIROMICS) Study of Early COPD Progression (SOURCE), a multicenter observational cohort study of younger individuals with a history of cigarette smoking and thus at-risk for, or with, early-stage COPD. Our overall objectives are to identify those who will develop COPD earlier in life, characterize them thoroughly, and by contrasting them to those not developing COPD, define mechanisms of disease progression. Methods/Discussion SOURCE utilizes the established SPIROMICS clinical network. Its goal is to enroll n=649 participants, ages 30-55 years, all races/ethnicities, with ≥10 pack-years cigarette smoking, in either Global initiative for chronic Obstructive Lung Disease (GOLD) groups 0-2 or with preserved ratio-impaired spirometry; and an additional n=40 never-smoker controls. Participants undergo baseline and 3-year follow-up visits, each including high-resolution computed tomography, respiratory oscillometry and spirometry (pre- and postbronchodilator administration), exhaled breath condensate (baseline only), and extensive biospecimen collection, including sputum induction. Symptoms, interim health care utilization, and exacerbations are captured every 6 months via follow-up phone calls. An embedded bronchoscopy substudy involving n=100 participants (including all never-smokers) will allow collection of lower airway samples for genetic, epigenetic, genomic, immunological, microbiome, mucin analyses, and basal cell culture. Conclusion SOURCE should provide novel insights into the natural history of lung disease in younger individuals with a smoking history, and its biological basis.
Collapse
Affiliation(s)
- Jeffrey L. Curtis
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States
- *Co-first authors
| | - Lori A. Bateman
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- *Co-first authors
| | - Susan Murray
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, United States
| | - David J. Couper
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Wassim W. Labaki
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Christine M. Freeman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States
| | - Stephanie A. Christenson
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of California San Francisco, San Francisco, California, United States
| | - Neil E. Alexis
- Division of Allergy, Immunology, and Infectious Disease, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Mehmet Kesimer
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Richard C. Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Robert J. Kaner
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York City, New York, United States
| | - Igor Barjaktarevic
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Christopher B. Cooper
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Eric A. Hoffman
- Department of Radiology, University of Iowa, Iowa City, Iowa, United States
| | - R. Graham Barr
- Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York City, New York, United States
| | - Eugene R. Bleecker
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Scottsdale, Arizona, United States
| | - Russell P. Bowler
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, United States
| | - Alejandro Comellas
- Department of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Mark T. Dransfield
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Michael B. Freedman
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Nadia N. Hansel
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
| | - Jerry A. Krishnan
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States
| | - Deborah A. Meyers
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, United States
| | - Jill Ohar
- Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University, Winston-Salem, North Carolina, United States
| | - Wanda K. O'Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Victor E. Ortega
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Scottsdale, Arizona, United States
| | - Robert Paine III
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, United States
| | - Stephen P. Peters
- Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University, Winston-Salem, North Carolina, United States
| | - Benjamin M. Smith
- Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York City, New York, United States
| | | | - J. Michael Wells
- Department of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Prescott G. Woodruff
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of California San Francisco, San Francisco, California, United States
| | - MeiLan K. Han
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
- **Co-senior authors
| | - Fernando J. Martinez
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York City, New York, United States
- **Co-senior authors
| | - for the SOURCE Investigators
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States
- Collaborative Studies Coordinating Center, Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, United States
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, University of California San Francisco, San Francisco, California, United States
- Division of Allergy, Immunology, and Infectious Disease, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York City, New York, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California Los Angeles, Los Angeles, California, United States
- Department of Radiology, University of Iowa, Iowa City, Iowa, United States
- Department of Medicine and Department of Epidemiology, Columbia University Medical Center, New York City, New York, United States
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Scottsdale, Arizona, United States
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, United States
- Department of Medicine, University of Iowa, Iowa City, Iowa, United States
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois, United States
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, United States
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, United States
- Section of Pulmonary, Critical Care, Allergy and Immunologic Diseases, Wake Forest University, Winston-Salem, North Carolina, United States
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, United States
- National Heart and Lung Institute, Imperial College, London, United Kingdom
- *Co-first authors
- **Co-senior authors
| |
Collapse
|
10
|
Choi JY, Rhee CK. It is high time to discard a cut-off of 0.70 in the diagnosis of COPD. Expert Rev Respir Med 2024; 18:709-719. [PMID: 39189795 DOI: 10.1080/17476348.2024.2397480] [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: 01/10/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) has traditionally been diagnosed based on the criterion of an FEV1/FVC <0.70. However, this definition has limitations as it may only detect patients with later-stage disease, when pathologic changes have become irreversible. Consequently, it potentially omits individuals with early-stage disease, in whom the pathologic changes could be delayed or reversed. AREAS COVERED This narrative review summarizes recent evidence regarding early-stage COPD, which may not fulfill the spirometric criteria but nonetheless exhibits features of COPD or is at risk of future COPD progression. EXPERT OPINION A comprehensive approach, including symptoms assessment, various physiologic tests, and radiologic features, is required to diagnose COPD. This approach is necessary to identify currently underdiagnosed patients and to halt disease progression in at- risk patients.
Collapse
Affiliation(s)
- Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chin Kook Rhee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
11
|
Çolak Y, Lange P, Vestbo J, Nordestgaard BG, Afzal S. Susceptible Young Adults and Development of Chronic Obstructive Pulmonary Disease Later in Life. Am J Respir Crit Care Med 2024; 210:607-617. [PMID: 38364200 DOI: 10.1164/rccm.202308-1452oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 02/16/2024] [Indexed: 02/18/2024] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) has its origin in early life, and the Global Initiative for Chronic Obstructive Lung Disease (GOLD) proposes a predisease state termed "pre-COPD." Objectives: We tested the hypothesis that susceptible young adults identified with chronic bronchitis and subtle lung function impairment will develop COPD later in life. Methods: We followed random individuals without COPD ages 20-50 years from two population-based cohorts from different smoking eras-the Copenhagen General Population Study from 2003 (N = 5,497) and the Copenhagen City Heart Study from 1976-1978 (N = 2,609)-for 10 and 25 years, for the development of COPD (FEV1/FVC <0.70) and COPD GOLD Stages 2-4 (additionally, FEV1 <80% predicted). Measurements and Main Results: After 10 years, 28% developed COPD and 13% developed COPD GOLD Stages 2-4 in individuals susceptible to COPD, compared with 8% and 1% in those without any susceptibility to COPD. Correspondingly, after 25 years, 22% versus 13% developed COPD and 20% versus 8% developed COPD GOLD Stages 2-4. More than half of incident COPD cases developed from a susceptible state. Compared with those without susceptibility to COPD, multivariable-adjusted odds ratios in those susceptible to COPD were 3.42 (95% confidence interval: 2.78-4.21) for COPD and 10.1 (6.77-15.2) for COPD GOLD Stages 2-4 after 10 years and were 1.54 (1.23-1.93) and 2.12 (1.64-2.73) after 25 years. The ability of a COPD risk score-consisting of the state of susceptibility to COPD with smoking and asthma as risk factors-to predict COPD later in life was high. Conclusions: Our study suggests the existence of a predisease state of COPD, which can be used for early identification of susceptible individuals at risk for COPD later in life.
Collapse
Affiliation(s)
- Yunus Çolak
- Department of Respiratory Medicine
- The Copenhagen General Population Study, and
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, and
| | - Peter Lange
- Department of Respiratory Medicine
- The Copenhagen General Population Study, and
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, and
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark; and
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Børge G Nordestgaard
- The Copenhagen General Population Study, and
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, and
| | - Shoaib Afzal
- The Copenhagen General Population Study, and
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, and
| |
Collapse
|
12
|
Pajand Birjandi M, Ammous O, Kampo R, Stanzel S, Wollsching-Strobel M, Mathes T. Care pathways versus usual care for chronic obstructive pulmonary disease (COPD). Cochrane Database Syst Rev 2024; 8:CD015800. [PMID: 39140370 PMCID: PMC11323265 DOI: 10.1002/14651858.cd015800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To assess the effects of care pathways (CPs) compared to usual care/no CPs for people with chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
| | - Omar Ammous
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Regina Kampo
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| | - Sarah Stanzel
- Department of Pneumology, Cologne Merheim Hospital, Kliniken der Stadt Köln gGmbH, Witten/Herdecke University, Cologne, Germany
| | - Maximilian Wollsching-Strobel
- Department of Pneumology, Cologne Merheim Hospital, Kliniken der Stadt Köln gGmbH, Witten/Herdecke University, Cologne, Germany
| | - Tim Mathes
- Department of Medical Statistics, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
13
|
Abdalla M, Elalami R, Cho MH, O’Connor GT, Rice M, Horowitz M, Akhoundi N, Yen A, Kalhan R, Diaz AA. Airway Mucus Plugs in Community-Living Adults: A Study Protocol. JOURNAL OF CLINICAL & EXPERIMENTAL PATHOLOGY 2024; 14:492. [PMID: 39360267 PMCID: PMC11446186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Introduction Mucus pathology plays a critical role in airway diseases like Chronic Bronchitis (CB) and Chronic Obstructive Pulmonary Disease (COPD). Up to 32% of community-living persons report clinical manifestations of mucus pathology (e.g., cough and sputum production). However, airway mucus pathology has not been systematically studied in community-living individuals. In this study, we will use an objective, reproducible assessment of mucus pathology on chest Computed Tomography (CT) scans from community-living individuals participating in the Coronary Artery Risk Development in Young Adults (CARDIA) and Framingham Heart Study (FHS) cohorts. Methods and analysis We will determine the clinical relevance of CT-based mucus plugs and modifiable and genetic risk and protective factors associated with this process. We will evaluate the associations of mucus plugs with lung function, respiratory symptoms, and chronic bronchitis and examine whether 5-yr. persistent CT-based mucus plugs are associated with the decline in FEV1 and future COPD. Also, we will assess whether modifiable factors, including air pollution and marijuana smoking are associated with increased odds of CT-based mucus plugs and whether cardiorespiratory fitness is related in an opposing manner. Finally, we will determine genetic resilience/susceptibility to mucus pathology. We will use CT data from the FHS and CARDIA cohorts and genome-wide sequencing data from the TOPMed initiative to identify common and rare variants associated with CT-based mucus plugging. Ethics and dissemination The Mass General Brigham Institutional Review Board approved the study. Findings will be disseminated through peer-reviewed journals and at professional conferences. Conclusion Determine whether the presence of CT-based mucus plugs is associated with lung health impairment, including reduced FEV1, more respiratory symptoms, and asthma. Identify modifiable risk and protective factors, such as pollution, exercise, smoking, and fitness that are associated with mucus plugs.
Collapse
Affiliation(s)
- Maya Abdalla
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Rim Elalami
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Michael H Cho
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - George T O’Connor
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
- The National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, Massachusetts, USA
| | - Mary Rice
- Division of Pulmonary, Sleep and Critical Care Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Michael Horowitz
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Neda Akhoundi
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Andrew Yen
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Ravi Kalhan
- Northwestern University Feinberg School of Medicine, 1700 W. Van Buren St, Ste. 470, 60612, Chicago, IL, USA
| | - Alejandro A. Diaz
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care, Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Abdalla M, Elalami R, Cho MH, O'Connor GT, Rice M, Horowitz M, Akhoundi N, Yen A, Kalhan R, Diaz AA. Airway Mucus Plugs in Community-Living Adults: A Study Protocol. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.15.24307439. [PMID: 38798504 PMCID: PMC11118634 DOI: 10.1101/2024.05.15.24307439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Introduction Mucus pathology plays a critical role in airway diseases like chronic bronchitis (CB) and chronic obstructive pulmonary disease (COPD). Up to 32% of community-living persons report clinical manifestations of mucus pathology (e.g., cough and sputum production). However, airway mucus pathology has not been systematically studied in community-living individuals. In this study, we will use an objective, reproducible assessment of mucus pathology on chest computed tomography (CT) scans from community-living individuals participating in the Coronary Artery Risk Development in Young Adults (CARDIA) and Framingham Heart Study (FHS) cohorts. Methods and analysis We will determine the clinical relevance of CT-based mucus plugs and modifiable and genetic risk and protective factors associated with this process. We will evaluate the associations of mucus plugs with lung function, respiratory symptoms, and chronic bronchitis and examine whether 5-yr. persistent CT-based mucus plugs are associated with the decline in FEV1 and future COPD. Also, we will assess whether modifiable factors, including air pollution and marijuana smoking are associated with increased odds of CT-based mucus plugs and whether cardiorespiratory fitness is related in an opposing manner. Finally, we will determine genetic resilience/susceptibility to mucus pathology. We will use CT data from the FHS and CARDIA cohorts and genome-wide sequencing data from the TOPMed initiative to identify common and rare variants associated with CT-based mucus plugging. Ethics and Dissemination The Mass General Brigham Institutional Review Board approved the study. Findings will be disseminated through peer-reviewed journals and at professional conferences.
Collapse
Affiliation(s)
- Maya Abdalla
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Rim Elalami
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Boston, MA
| | - Michael H Cho
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA
| | - George T O'Connor
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- The National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, Massachusetts, United States of America
| | - Mary Rice
- Division of Pulmonary, Sleep and Critical Care Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Michael Horowitz
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Neda Akhoundi
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Andrew Yen
- Department of Radiology, University of California, San Diego, 9452 Medical Center Dr, 4th Floor, La Jolla, CA 92037
| | - Ravi Kalhan
- Northwestern University Feinberg School of Medicine, 1700 W. Van Buren St, Ste. 470, 60612, Chicago, IL, USA
| | - Alejandro A Diaz
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Ritchie AI, Donaldson GC, Hoffman EA, Allinson JP, Bloom CI, Bolton CE, Choudhury G, Gerard SE, Guo J, Alves-Moreira L, McGarvey L, Sapey E, Stockley RA, Yip KP, Singh D, Wilkinson T, Fageras M, Ostridge K, Jöns O, Bucchioni E, Compton CH, Jones P, Mezzi K, Vestbo J, Calverley PMA, Wedzicha JA. Structural Predictors of Lung Function Decline in Young Smokers with Normal Spirometry. Am J Respir Crit Care Med 2024; 209:1208-1218. [PMID: 38175920 PMCID: PMC11146542 DOI: 10.1164/rccm.202307-1203oc] [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: 07/14/2023] [Accepted: 01/04/2024] [Indexed: 01/06/2024] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) due to tobacco smoking commonly presents when extensive lung damage has occurred. Objectives: We hypothesized that structural change would be detected early in the natural history of COPD and would relate to loss of lung function with time. Methods: We recruited 431 current smokers (median age, 39 yr; 16 pack-years smoked) and recorded symptoms using the COPD Assessment Test (CAT), spirometry, and quantitative thoracic computed tomography (QCT) scans at study entry. These scan results were compared with those from 67 never-smoking control subjects. Three hundred sixty-eight participants were followed every six months with measurement of postbronchodilator spirometry for a median of 32 months. The rate of FEV1 decline, adjusted for current smoking status, age, and sex, was related to the initial QCT appearances and symptoms, measured using the CAT. Measurements and Main Results: There were no material differences in demography or subjective CT appearances between the young smokers and control subjects, but 55.7% of the former had CAT scores greater than 10, and 24.2% reported chronic bronchitis. QCT assessments of disease probability-defined functional small airway disease, ground-glass opacification, bronchovascular prominence, and ratio of small blood vessel volume to total pulmonary vessel volume were increased compared with control subjects and were all associated with a faster FEV1 decline, as was a higher CAT score. Conclusions: Radiological abnormalities on CT are already established in young smokers with normal lung function and are associated with FEV1 loss independently of the impact of symptoms. Structural abnormalities are present early in the natural history of COPD and are markers of disease progression. Clinical trial registered with www.clinicaltrials.gov (NCT03480347).
Collapse
Affiliation(s)
- Andrew I. Ritchie
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- AstraZeneca, Cambridge, United Kingdom
| | - Gavin C. Donaldson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Eric A. Hoffman
- Department of Radiology and
- Roy J. Carver Department of Biomedical Engineering, Medicine and Biomedical Engineering, University of Iowa, Iowa City, Iowa
| | - James P. Allinson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Royal Brompton Hospital, London, United Kingdom
| | - Chloe I. Bloom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Charlotte E. Bolton
- NIHR Nottingham Biomedical Research Centre
- Centre for Respiratory Research, NIHR Nottingham, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Gourab Choudhury
- ELEGI and COLT Laboratories, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Sarah E. Gerard
- Roy J. Carver Department of Biomedical Engineering, Medicine and Biomedical Engineering, University of Iowa, Iowa City, Iowa
| | | | - Luana Alves-Moreira
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Lorcan McGarvey
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, United Kingdom
- Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Robert A. Stockley
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - K. P. Yip
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Tom Wilkinson
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- National Institute for Health and Care Research Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | | | - Kristoffer Ostridge
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- AstraZeneca, Gothenburg, Sweden
| | - Olaf Jöns
- Boehringer Ingelheim International GmbH, Ingelheim am Rhein, Germany
| | | | | | - Paul Jones
- GlaxoSmithKline, Brentford, United Kingdom
| | | | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Peter M. A. Calverley
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jadwiga A. Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
16
|
Pistenmaa CL, Washko GR. BEACON: A Missing Piece of the Puzzle for Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2024; 209:1177-1178. [PMID: 38330311 PMCID: PMC11146538 DOI: 10.1164/rccm.202401-0144ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/08/2024] [Indexed: 02/10/2024] Open
Affiliation(s)
- Carrie L Pistenmaa
- Department of Medicine Brigham and Women's Hospital Boston, Massachusetts
| | - George R Washko
- Department of Medicine Brigham and Women's Hospital Boston, Massachusetts
| |
Collapse
|
17
|
Papi A, Alfano F, Bigoni T, Mancini L, Mawass A, Baraldi F, Aljama C, Contoli M, Miravitlles M. N-acetylcysteine Treatment in Chronic Obstructive Pulmonary Disease (COPD) and Chronic Bronchitis/Pre-COPD: Distinct Meta-analyses. Arch Bronconeumol 2024; 60:269-278. [PMID: 38555190 DOI: 10.1016/j.arbres.2024.03.010] [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: 12/22/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/02/2024]
Abstract
INTRODUCTION N-acetylcysteine (NAC) is a mucolytic agent with antioxidant properties. Oxidative stress is a key pathogenic mechanism in chronic respiratory conditions such as COPD and chronic bronchitis (CB). In these meta-analyses we investigated the efficacy of NAC in subjects with COPD or CB, the latter being a potential pre-COPD condition (CB/pre-COPD). METHODS The meta-analyses were conducted according to PRISMA guidelines. Exacerbations were assessed using total number of exacerbations. Improvement in patients' respiratory symptoms and/or patients quality of life (QoL) were measured by validated tools or assessed at the end of the study. RESULTS Twenty studies were included, of which seven evaluated NAC in patients with symptoms of CB/pre-COPD as entry criterion. NAC treated patients showed a significant reduction of the incidence of exacerbations as compared to placebo both in COPD (IRR=0.76; 95% confidence interval (CI) 0.59-0.99) and CB/pre-COPD (IRR=0.81; 95% CI 0.69-0.95). Sensitivity analyses in studies with duration higher than 5 months, confirmed the overall results. CB/pre-COPD patients treated with NAC were significantly more likely to experience an improvement in symptoms and/or QoL compared to placebo (odds ratio (OR)=3.47; 95% CI 1.92-6.26). A similar trend was observed in the few COPD studies evaluable. Sensitivity analyses showed a significant association of NAC with improvement in symptoms and/or QoL both in CB/pre-COPD and COPD patients. CONCLUSIONS These findings provide novel data of NAC on the improvement in symptoms and QoL in addition to prevention of exacerbations in COPD and CB/pre-COPD. PROSPERO registry no. CRD42023468154.
Collapse
Affiliation(s)
- Alberto Papi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'Anna University Hospital, Ferrara, Italy.
| | - Franco Alfano
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'Anna University Hospital, Ferrara, Italy
| | - Tommaso Bigoni
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'Anna University Hospital, Ferrara, Italy
| | | | | | - Federico Baraldi
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'Anna University Hospital, Ferrara, Italy
| | - Cristina Aljama
- Pneumology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Marco Contoli
- Department of Translational Medicine, University of Ferrara Medical School, University of Ferrara, Sant'Anna University Hospital, Ferrara, Italy
| | - Marc Miravitlles
- Pneumology Department, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain
| |
Collapse
|
18
|
Yoshida A, Kai C, Futamura H, Oochi K, Kondo S, Sato I, Kasai S. Spirometry test values can be estimated from a single chest radiograph. Front Med (Lausanne) 2024; 11:1335958. [PMID: 38510449 PMCID: PMC10953498 DOI: 10.3389/fmed.2024.1335958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/23/2024] [Indexed: 03/22/2024] Open
Abstract
Introduction Physical measurements of expiratory flow volume and speed can be obtained using spirometry. These measurements have been used for the diagnosis and risk assessment of chronic obstructive pulmonary disease and play a crucial role in delivering early care. However, spirometry is not performed frequently in routine clinical practice, thereby hindering the early detection of pulmonary function impairment. Chest radiographs (CXRs), though acquired frequently, are not used to measure pulmonary functional information. This study aimed to evaluate whether spirometry parameters can be estimated accurately from single frontal CXR without image findings using deep learning. Methods Forced vital capacity (FVC), forced expiratory volume in 1 s (FEV1), and FEV1/FVC as spirometry measurements as well as the corresponding chest radiographs of 11,837 participants were used in this study. The data were randomly allocated to the training, validation, and evaluation datasets at an 8:1:1 ratio. A deep learning network was pretrained using ImageNet. The input and output information were CXRs and spirometry test values, respectively. The training and evaluation of the deep learning network were performed separately for each parameter. The mean absolute error rate (MAPE) and Pearson's correlation coefficient (r) were used as the evaluation indices. Results The MAPEs between the spirometry measurements and AI estimates for FVC, FEV1 and FEV1/FVC were 7.59% (r = 0.910), 9.06% (r = 0.879) and 5.21% (r = 0.522), respectively. A strong positive correlation was observed between the measured and predicted indices of FVC and FEV1. The average accuracy of >90% was obtained in each estimation of spirometry indices. Bland-Altman analysis revealed good agreement between the estimated and measured values for FVC and FEV1. Discussion Frontal CXRs contain information related to pulmonary function, and AI estimation performed using frontal CXRs without image findings could accurately estimate spirometry values. The network proposed for estimating pulmonary function in this study could serve as a recommendation for performing spirometry or as an alternative method, suggesting its utility.
Collapse
Affiliation(s)
- Akifumi Yoshida
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, Niigata, Japan
| | - Chiharu Kai
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, Niigata, Japan
- Major in Health and Welfare, Graduate School of Niigata University of Health and Welfare, Niigata, Japan
| | | | | | - Satoshi Kondo
- Graduate School of Engineering, Muroran Institute of Technology, Muroran, Japan
| | - Ikumi Sato
- Major in Health and Welfare, Graduate School of Niigata University of Health and Welfare, Niigata, Japan
- Department of Nursing, Faculty of Nursing, Niigata University of Health and Welfare, Niigata, Japan
| | - Satoshi Kasai
- Department of Radiological Technology, Faculty of Medical Technology, Niigata University of Health and Welfare, Niigata, Japan
| |
Collapse
|
19
|
Fan J, Fang L, Cong S, Zhang Y, Jiang X, Wang N, Chen Y. Potential pre-COPD indicators in association with COPD development and COPD prediction models in Chinese: a prospective cohort study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 44:100984. [PMID: 38186582 PMCID: PMC10770747 DOI: 10.1016/j.lanwpc.2023.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024]
Abstract
Background Lung injury might take place before chronic obstructive pulmonary disease (COPD) occurs. A clearer definition of "pre-COPD" based on the effects of potential indicators on increasing risk of COPD development and a prediction model involving them are lacking. Methods A total of 3526 Chinese residents without COPD aged 40 years or older derived from the national cross-sectional survey of COPD surveillance in 2014-2015 were followed up for a mean of 3.59 years. We examined the associations of chronic bronchitis, preserved ratio impaired spirometry (PRISm), low peak expiratory flow (PEF), spirometric small airway dysfunction (sSAD), low maximal mid-expiratory flow (MMEF), low forced expiratory flow 50% of pulmonary volume (FEF50), and low FEF75 with subsequent COPD and constructed a prediction model with LASSO-Cox regression. Findings 235 subjects in the cohort developed COPD during the follow-up. Subjects with PRISm, low PEF, sSAD, low MMEF, low FEF50, and low FEF75 had an increased risk of developing COPD (adjusted hazard ratio [HR] ranging from 1.57 to 3.01). Only chronic bronchitis (HR 2.84 [95% CI 1.38-5.84] and 2.94 [1.43-6.04]) and sSAD/low MMEF (HR 2.74 [2.07-3.61] and 2.38 [1.65-3.43]) showed effects independent of the other indicators and their concurrence had the strongest effect (HR 5.89 and 4.80). The prediction model including age, sex, low MMEF, low FEF50, and indoor exposure to biomass had good performance both internally and temporally. The corrected C-index was 0.77 (0.72-0.81) for discrimination in internal validation. For temporal validation, the area under the receiver operating characteristic curve was 0.73 (0.63-0.83). Good calibration was indicated in plot for internal validation and by Hosmer-Lemeshow test for temporal validation. Interpretation Individuals with concurrent chronic bronchitis and sSAD/low MMEF indicating pre-COPD optimally require more high attention from physicians. Our prediction model could serve as a multi-dimension tool to predict COPD comprehensively. Funding The Ministry of Finance and the Ministry of Science and Technology of the People's Republic of China and the National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Jing Fan
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing 100191, China
| | - Liwen Fang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Shu Cong
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Yang Zhang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Xiao Jiang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Ning Wang
- National Center for Chronic and Non-communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 27 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, 49 Huayuan North Road, Haidian District, Beijing 100191, China
| |
Collapse
|
20
|
Hou Y, Wu F, Fan H, Li H, Hao B, Deng Z, Lu X, Zhou Y, Ran P. Association of non-obstructive dyspnoea with all-cause mortality and incident chronic obstructive pulmonary disease: a systematic literature review and meta-analysis. BMJ Open Respir Res 2024; 11:e001933. [PMID: 38395457 PMCID: PMC10895236 DOI: 10.1136/bmjresp-2023-001933] [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: 07/05/2023] [Accepted: 01/19/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Controversy exists regarding the association between non-obstructive dyspnoea and the future development of chronic obstructive pulmonary disease (COPD) and mortality. Therefore, we aimed to evaluate the association of non-obstructive dyspnoea with mortality and incident COPD in adults. METHODS We searched PubMed, Embase, and Web of Science to identify studies published from inception to 13 May 2023. Eligibility screening, data extraction, and quality assessment of the retrieved articles were conducted independently by two reviewers. Studies were included if they were original articles comparing incident COPD and all-cause mortality between individuals with normal lung function with and without dyspnoea. The primary outcomes were incident COPD and all-cause mortality. The secondary outcome was respiratory disease-related mortality. We used the random-effects model to calculate pooled estimates and corresponding 95% confidence interval (CI). Heterogeneity was determined using the I² statistic. RESULTS Of 6486 studies, 8 studies involving 100 758 individuals fulfilled the inclusion and exclusion criteria and were included in the study. Compared with individuals without non-obstructive dyspnoea, individuals with non-obstructive dyspnoea had an increased risk of incident COPD (relative risk: 1.41, 95% CI: 1.08 to 1.83), and moderate heterogeneity was found (p=0.079, I2=52.2%). Individuals with non-obstructive dyspnoea had a higher risk of all-cause mortality (hazard ratio: 1.21, 95% CI: 1.14 to 1.28, I2=0.0%) and respiratory disease-related mortality (hazard ratio: 1.52, 95% CI: 1.14 to 2.02, I2=0.0%) than those without. CONCLUSIONS Individuals with non-obstructive dyspnoea are at a higher risk of incident COPD and all-cause mortality than individuals without dyspnoea. Further research should investigate whether these high-risk adults may benefit from risk management and early therapeutic intervention. PROSPERO REGISTRATION NUMBER CRD42023395192.
Collapse
Affiliation(s)
- Yuyan Hou
- Jiaying University, Meizhou, Guangdong, China
| | - Fan Wu
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Huanhuan Fan
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haiqing Li
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Binwei Hao
- Department of Pulmonary and Critical Care Medicine, Shanxi Bethune Hospital Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhishan Deng
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Lu
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yumin Zhou
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Pixin Ran
- State Key Laboratory of Respiratory Disease & Guangzhou Institute of Respiratory Health & National Clinical Research Center for Respiratory Disease & National Center for Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
21
|
Papi A, Faner R, Pavord I, Baraldi F, McDonald VM, Thomas M, Miravitlles M, Roche N, Agustí A. From treatable traits to GETomics in airway disease: moving towards clinical practice. Eur Respir Rev 2024; 33:230143. [PMID: 38232989 DOI: 10.1183/16000617.0143-2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/24/2023] [Indexed: 01/19/2024] Open
Abstract
The treatable traits approach represents a strategy for patient management. It is based on the identification of characteristics susceptible to treatments or predictive of treatment response in each individual patient. With the objective of accelerating progress in research and clinical practice relating to such a treatable traits approach, the Portraits event was convened in Barcelona, Spain, in November 2022. Here, while reporting the key concepts that emerged from the discussions during the meeting, we review the current state of the art related to treatable traits and chronic respiratory diseases management, and we describe the possible actions that clinicians can take in clinical practice to implement the treatable traits framework. Furthermore, we explore the new concept of GETomics and the new models of research in the field of COPD.
Collapse
Affiliation(s)
- Alberto Papi
- Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Rosa Faner
- University of Barcelona, Biomedicine Department, FCRB-IDIBAPS, Centro de Investigación Biomedica en Red M.P. (CIBER), Barcelona, Spain
| | - Ian Pavord
- Respiratory Medicine Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Vanessa M McDonald
- School of Nursing and Midwifery, The University of Newcastle, NHMRC Centre of Excellence in Asthma Treatable Traits, Hunter Medical Research Institute Asthma and Breathing Research Programme and Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Mike Thomas
- Primary Care and Population Sciences, University of Southampton, Southampton, UK
| | - Marc Miravitlles
- Pneumology Department Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Nicholas Roche
- Respiratory Medicine Department, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, AP-HP and Université Paris Cité, Paris, France
| | - Alvar Agustí
- University of Barcelona, Hospital Clinic, IDIBAPS and CIBERES, Barcelona, Spain
- Pulmonary Service, Respiratory Institute, Clinic Barcelona, Barcelona, Spain
| |
Collapse
|
22
|
Gan Q, Yang K, Wu Y, Wang J, Zhang H, Zhang N, Wu K. Icenticaftor, a Novel Treatment for Chronic Obstructive Pulmonary Disease, Needs Further Verification of Its Effectiveness. Am J Respir Crit Care Med 2023; 208:1140-1141. [PMID: 37748186 PMCID: PMC10867933 DOI: 10.1164/rccm.202307-1268le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Affiliation(s)
- Qiming Gan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Kai Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yanjuan Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jingcun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Haojie Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Kang Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, National Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
23
|
Tesfaigzi Y, Curtis JL, Petrache I, Polverino F, Kheradmand F, Adcock IM, Rennard SI. Does Chronic Obstructive Pulmonary Disease Originate from Different Cell Types? Am J Respir Cell Mol Biol 2023; 69:500-507. [PMID: 37584669 PMCID: PMC10633838 DOI: 10.1165/rcmb.2023-0175ps] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/16/2023] [Indexed: 08/17/2023] Open
Abstract
The onset of chronic obstructive pulmonary disease (COPD) is heterogeneous, and current approaches to define distinct disease phenotypes are lacking. In addition to clinical methodologies, subtyping COPD has also been challenged by the reliance on human lung samples from late-stage diseases. Different COPD phenotypes may be initiated from the susceptibility of different cell types to cigarette smoke, environmental pollution, and infections at early stages that ultimately converge at later stages in airway remodeling and destruction of the alveoli when the disease is diagnosed. This perspective provides discussion points on how studies to date define different cell types of the lung that can initiate COPD pathogenesis, focusing on the susceptibility of macrophages, T and B cells, mast cells, dendritic cells, endothelial cells, and airway epithelial cells. Additional cell types, including fibroblasts, smooth muscle cells, neuronal cells, and other rare cell types not covered here, may also play a role in orchestrating COPD. Here, we discuss current knowledge gaps, such as which cell types drive distinct disease phenotypes and/or stages of the disease and which cells are primarily affected by the genetic variants identified by whole genome-wide association studies. Applying new technologies that interrogate the functional role of a specific cell type or a combination of cell types as well as single-cell transcriptomics and proteomic approaches are creating new opportunities to understand and clarify the pathophysiology and thereby the clinical heterogeneity of COPD.
Collapse
Affiliation(s)
- Yohannes Tesfaigzi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey L. Curtis
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
- Division of Pulmonary and Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Irina Petrache
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado
- University of Colorado, Denver, Colorado
| | - Francesca Polverino
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Farrah Kheradmand
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, Baylor University, Houston, Texas
| | - Ian M. Adcock
- Department of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom; and
| | - Stephen I. Rennard
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
24
|
Divo MJ, Liu C, Polverino F, Castaldi PJ, Celli BR, Tesfaigzi Y. From pre-COPD to COPD: a Simple, Low cost and easy to IMplement (SLIM) risk calculator. Eur Respir J 2023; 62:2300806. [PMID: 37678951 PMCID: PMC10533946 DOI: 10.1183/13993003.00806-2023] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/09/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND The lifetime risk of developing clinical COPD among smokers ranges from 13% to 22%. Identifying at-risk individuals who will develop overt disease in a reasonable timeframe may allow for early intervention. We hypothesised that readily available clinical and physiological variables could help identify ever-smokers at higher risk of developing chronic airflow limitation (CAL). METHODS Among 2273 Lovelace Smokers' Cohort (LSC) participants, we included 677 (mean age 54 years) with normal spirometry at baseline and a minimum of three spirometries, each 1 year apart. Repeated spirometric measurements were used to determine incident CAL. Using logistic regression, demographics, anthropometrics, smoking history, modified Medical Research Council dyspnoea scale, St George's Respiratory Questionnaire, comorbidities and spirometry, we related variables obtained at baseline to incident CAL as defined by the Global Initiative for Chronic Obstructive Lung Disease and lower limit of normal criteria. The predictive model derived from the LSC was validated in subjects from the COPDGene study. RESULTS Over 6.3 years, the incidence of CAL was 26 cases per 1000 person-years. The strongest independent predictors were forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) <0.75, having smoked ≥30 pack-years, body mass index (BMI) ≤25 kg·m2 and symptoms of chronic bronchitis. Having all four predictors increased the risk of developing CAL over 6 years to 85% (area under the receiver operating characteristic curve (AUC ROC) 0.84, 95% CI 0.81-0.89). The prediction model showed similar results when applied to subjects in the COPDGene study with a follow-up period of 10 years (AUC ROC 0.77, 95% CI 0.72-0.81). CONCLUSION In middle-aged ever-smokers, a simple predictive model with FEV1/FVC, smoking history, BMI and chronic bronchitis helps identify subjects at high risk of developing CAL.
Collapse
Affiliation(s)
- Miguel J Divo
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Congjian Liu
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca Polverino
- Pulmonary and Critical Care Medicine, Department of Medicine, Baylor College of Medicine Houston, Houston, TX, USA
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- General Medicine and Primary Care, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bartolome R Celli
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- B.R. Celli and Y. Tesfaigzi are senior authors and contributed equally to this study and manuscript
| | - Yohannes Tesfaigzi
- Pulmonary and Critical Care Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- B.R. Celli and Y. Tesfaigzi are senior authors and contributed equally to this study and manuscript
| |
Collapse
|
25
|
Liu Z, Pan H, Liu B, Li L, Yang H, Shen T. Environmental and occupational risk factors for COPD and its prevalence among miners worldwide: a Mendelian randomization and meta-analysis study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:97545-97561. [PMID: 37592069 DOI: 10.1007/s11356-023-29269-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death after cardiovascular disease and stroke, and its incidence is associated with genetic, environmental, and occupational factors. Miner is high-risk population for COPD, but the global prevalence of COPD in this group is inaccurate. In this study, the environmental and occupational risk factors for COPD were explored comprehensively with a two-sample Mendelian randomization study by combining genome-wide association data from two large global sample sizes of publicly available databases, UK Biobank (n = 503,317) and FinnGen (n = 193,638), as well as the prevalence of COPD among miners was investigated with meta-analysis followed a random-effects model including seven studies (16,033 miners in total). This study found that asthma, smoking, shift work, and workplace dust exposure may increase an individual's risk of COPD. The pooled prevalence of COPD among miners globally was 12% (95% CI: 8%, 18%), with higher prevalence of COPD among ex-smokers and dust-exposed individuals, and was significantly influenced by the method of diagnosis. Our findings suggest that there is currently a lack of practical criteria for diagnosing COPD in the physical examination and screening of miners. The actual prevalence of COPD may be underestimated due to the healthy worker effect and the phenomenon of job switching, and appropriate policies should be favored in the future to reduce the risk of COPD in miner.
Collapse
Affiliation(s)
- Zikai Liu
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Haihong Pan
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Bin Liu
- Department of Medical Aspects of Specific Environments, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Lanlan Li
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Hongxu Yang
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Tong Shen
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
26
|
Martinez FJ, Criner GJ, Gessner C, Jandl M, Scherbovsky F, Shinkai M, Siler TM, Vogelmeier CF, Voves R, Wedzicha JA, Bartels C, Bottoli I, Byiers S, Cardenas P, Eckert JH, Gutzwiller FS, Knorr B, Kothari M, Parlikar R, Tanase AM, Franssen FM. Icenticaftor, a CFTR Potentiator, in COPD: A Multicenter, Parallel-Group, Double-Blind Clinical Trial. Am J Respir Crit Care Med 2023; 208:417-427. [PMID: 37411039 PMCID: PMC10449083 DOI: 10.1164/rccm.202303-0458oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Rationale: CFTR (cystic fibrosis transmembrane conductance regulator) dysfunction is associated with mucus accumulation and worsening chronic obstructive pulmonary disease (COPD) symptoms. Objectives: The aim of this phase IIb dose-finding study was to compare a CFTR potentiator, icenticaftor (QBW251), with placebo in patients with COPD and chronic bronchitis. Methods: Patients with COPD on triple therapy for at least three months were randomized to six treatment arms (icenticaftor 450, 300, 150, 75, or 25 mg or placebo twice daily [b.i.d.]) in a 24-week, multicenter, parallel-group, double-blind study. The primary endpoint was change from baseline in trough FEV1 after 12 weeks. Secondary endpoints included change from baseline in trough FEV1 and Evaluating Respiratory Symptoms in COPD (E-RS) total and cough and sputum scores after 24 weeks. Multiple comparison procedure-modeling was conducted to characterize dose-response relationship. Rescue medication use, exacerbations, and change in serum fibrinogen concentration after 24 weeks were assessed in exploratory and post hoc analyses, respectively. Measurements and Main Results: Nine hundred seventy-four patients were randomized. After 12 weeks of icenticaftor treatment, no dose-response relationship for change from baseline in trough FEV1 was observed; however, it was observed for E-RS cough and sputum score. A dose-response relationship was observed after 24 weeks for trough FEV1, E-RS cough and sputum and total scores, rescue medication use, and fibrinogen. A dose of 300 mg b.i.d. was consistently the most effective. Improvements for 300 mg b.i.d. versus placebo were also seen in pairwise comparisons of these endpoints. All treatments were well tolerated. Conclusions: The primary endpoint was negative, as icenticaftor did not improve trough FEV1 over 12 weeks. Although the findings must be interpreted with caution, icenticaftor improved trough FEV1; reduced cough, sputum, and rescue medication use; and lowered fibrinogen concentrations at 24 weeks. Clinical trial registered with www.clinicaltrials.gov (NCT04072887).
Collapse
Affiliation(s)
- Fernando J. Martinez
- Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine/NewYork-Presbyterian Hospital, New York, New York
| | - Gerard J. Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania
| | - Christian Gessner
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Margret Jandl
- Hamburger Institut für Therapieforschung GmbH, Hamburg, Germany
| | | | - Masaharu Shinkai
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo, Japan
| | | | - Claus F. Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, German Center for Lung Research, Marburg, Germany
| | - Robert Voves
- Private Practice, Bismarckstraße, Feldbach, Austria
| | - Jadwiga A. Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | | | - Pamela Cardenas
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | | | | - Barbara Knorr
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | | | | | | - Frits M.E. Franssen
- Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
27
|
Wedzicha JA. Causes of Death in Smokers: Implications for Chronic Obstructive Pulmonary Disease Management across Disease Severity. Am J Respir Crit Care Med 2023; 208:354-356. [PMID: 37429287 PMCID: PMC10449074 DOI: 10.1164/rccm.202306-1065ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 07/12/2023] Open
Affiliation(s)
- Jadwiga A Wedzicha
- National Heart and Lung Institute Imperial College London London, United Kingdom
| |
Collapse
|
28
|
Chan SMH, Brassington K, Almerdasi SA, Dobric A, De Luca SN, Coward‐Smith M, Wang H, Mou K, Akhtar A, Alateeq RA, Wang W, Seow HJ, Selemidis S, Bozinovski S, Vlahos R. Inhibition of oxidative stress by apocynin attenuated chronic obstructive pulmonary disease progression and vascular injury by cigarette smoke exposure. Br J Pharmacol 2023; 180:2018-2034. [PMID: 36908040 PMCID: PMC10953324 DOI: 10.1111/bph.16068] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/07/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Cardiovascular disease affects up to half of the patients with chronic obstructive pulmonary disease (COPD), exerting deleterious impact on health outcomes and survivability. Vascular endothelial dysfunction marks the onset of cardiovascular disease. The present study examined the effect of a potent NADPH Oxidase (NOX) inhibitor and free-radical scavenger, apocynin, on COPD-related cardiovascular disease. EXPERIMENTAL APPROACH Male BALB/c mice were exposed to either room air (Sham) or cigarette smoke (CS) generated from 9 cigarettes·day-1 , 5 days a week for up to 24 weeks with or without apocynin treatment (5 mg·kg-1 ·day-1 , intraperitoneal injection). KEY RESULTS Eight-weeks of apocynin treatment reduced airway neutrophil infiltration (by 42%) and completely preserved endothelial function and endothelial nitric oxide synthase (eNOS) availability against the oxidative insults of cigarette smoke exposure. These preservative effects were maintained up until the 24-week time point. 24-week of apocynin treatment markedly reduced airway inflammation (reduced infiltration of macrophage, neutrophil and lymphocyte), lung function decline (hyperinflation) and prevented airway collagen deposition by cigarette smoke exposure. CONCLUSION AND IMPLICATIONS Limiting NOX activity may slow COPD progression and lower cardiovascular disease risk, particularly when signs of oxidative stress become evident.
Collapse
Affiliation(s)
- Stanley M. H. Chan
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Kurt Brassington
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Suleman Abdullah Almerdasi
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Aleksandar Dobric
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Simone N. De Luca
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Madison Coward‐Smith
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Hao Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Kevin Mou
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Alina Akhtar
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Rana Abdullah Alateeq
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Wei Wang
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Huei Jiunn Seow
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Steven Bozinovski
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| | - Ross Vlahos
- Centre for Respiratory Science and Health, School of Health and Biomedical SciencesRMIT UniversityBundooraVictoria3083Australia
| |
Collapse
|
29
|
Chen D, Curtis JL, Chen Y. Twenty years of changes in the definition of early chronic obstructive pulmonary disease. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:84-93. [PMID: 39170827 PMCID: PMC11332824 DOI: 10.1016/j.pccm.2023.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Indexed: 08/23/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory airway disease that affects the quality of life of nearly one-tenth of the global population. Due to irreversible airflow obstruction and progressive lung function decline, COPD is characterized by high mortality and disability rates, which imposes a huge economic burden on society. In recent years, the importance of intervention in the early stage of COPD has been recognized and the concept of early COPD has been proposed. Identifying and intervening in individuals with early COPD, some of whom have few or no symptoms, might halt or reverse the progressive decline in lung function, improve the quality of life, and better their prognosis. However, understanding of early COPD is not yet well established, and there are no unified and feasible diagnostic criteria, which complicates clinical research. In this article, we review evolution of the definition of early COPD over the past 20 years, describe the changes in awareness of this concept, and propose future research directions.
Collapse
Affiliation(s)
- Dian Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Jeffrey L. Curtis
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48105, USA
- Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Yahong Chen
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Research center for Chronic Airway Diseases, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
30
|
Tyrrell J, Ghosh A, Manzo ND, Randell SH, Tarran R. Evaluation of chronic cigarette smoke exposure in human bronchial epithelial cultures. J Appl Toxicol 2023; 43:862-873. [PMID: 36594405 DOI: 10.1002/jat.4430] [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: 10/18/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
Cigarette smoke (CS) exposure induces both cytotoxicity and inflammation, and often causes COPD, a growing cause of morbidity and mortality. CS also inhibits the CFTR Cl- channel, leading to airway surface liquid dehydration, which is predicated to impair clearance of inhaled pathogens and toxicants. Numerous in vitro studies have been performed that utilize acute (≤24 h) CS exposures. However, CS exposure is typically chronic. We evaluated the feasibility of using British-American Tobacco (BAT)-designed CS exposure chambers for chronically exposing human bronchial epithelial cultures (HBECs) to CS. HBECs are polarized and contain mucosal and serosal sides. In vivo, inhaled CS interacts with mucosal membranes, and BAT chambers are designed to direct CS to HBEC mucosal surfaces while keeping CS away from serosal surfaces via a perfusion system. We found that serosal perfusion was absolutely required to maintain HBEC viability over time following chronic CS exposure. Indeed, with this system, we found that CS increased inflammation and mucin levels, while decreasing CFTR function. Without this serosal perfusion, CS was extremely toxic within 24 h. We therefore propose that 5- and 10-day CS exposures with serosal perfusion are suitable for measuring chronic CS exposure and can be used for monitoring new and emerging tobacco products.
Collapse
Affiliation(s)
- Jean Tyrrell
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Arunava Ghosh
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Nicholas D Manzo
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Robert Tarran
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Abozid H, Kirby M, Nasir N, Hartl S, Breyer-Kohansal R, Breyer MK, Burghuber OC, Bourbeau J, Wouters EFM, Tan W. CT airway remodelling and chronic cough. BMJ Open Respir Res 2023; 10:10/1/e001462. [PMID: 37173074 DOI: 10.1136/bmjresp-2022-001462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
RATIONALE Structural airway changes related to chronic cough (CC) are described in the literature, but so far reported data are rare and non-conclusive. Furthermore, they derive mainly from cohorts with small sample sizes. Advanced CT imaging not only allows airway abnormalities to be quantified, but also to count the number of visible airways. The current study evaluates these airway abnormalities in CC and assesses the contribution of CC in addition to CT findings on the progression of airflow limitation, defined as a decline in forced expiratory volume in 1 s (FEV1) over time. METHODS A total of 1183 males and females aged ≥40 years with thoracic CT scans and valid spirometry from Canadian Obstructive Lung Disease, a Canadian multicentre, population-based study has been included in this analysis. Participants were stratified into 286 never-smokers, 297 ever-smokers with normal lung function and 600 with chronic obstructive pulmonary disease (COPD) of different severity grades. Imaging parameters analyses included total airway count (TAC), airway wall thickness, emphysema as well as parameters for functional small airway disease quantification. RESULTS Irrespective of COPD presence, CC was not related to specific airway and lung structure features. Independent of TAC and emphysema score, CC was highly associated with FEV1 decline over time in the entire study population, particularly in ever-smokers (p<0.0001). CONCLUSION The absence of specific structural CT features independently from COPD presence indicate that other underlying mechanisms are contributing to the symptomatology of CC. On top of derived CT parameters, CC seems to be independently associated with FEV1 decline. TRIAL REGISTRATION NUMBER NCT00920348.
Collapse
Affiliation(s)
- Hazim Abozid
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, Vienna, Austria
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Miranda Kirby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Neha Nasir
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Sylvia Hartl
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, Vienna Healthcare Group, Vienna, Austria
| | - Robab Breyer-Kohansal
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, Vienna Healthcare Group, Vienna, Austria
| | - Marie-Kathrin Breyer
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Department of Respiratory and Pulmonary Diseases, Clinic Penzing, Vienna Healthcare Group, Vienna, Austria
| | - Otto C Burghuber
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Faculty for Medicine, Sigmund Freud University, Vienna, Austria
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, Research Institute, McGill University, Montreal, Québec, Canada
| | - Emiel F M Wouters
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
- Maastricht University Medical Center, Maastricht, The Netherlands
| | - Wan Tan
- Centre for Heart Lung Innovation, University of British Columbia, St Pauls's Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Cosío BG, Casanova C, Soler-Cataluña JJ, Soriano JB, García-Río F, de Lucas P, Alfageme I, Rodríguez González-Moro JM, Sánchez G, Ancochea J, Miravitlles M. Unravelling young COPD and pre-COPD in the general population. ERJ Open Res 2023; 9:00334-2022. [PMID: 36814553 PMCID: PMC9940715 DOI: 10.1183/23120541.00334-2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/24/2022] [Indexed: 11/05/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is commonly diagnosed when the airflow limitation is well established and symptomatic. We aimed to identify individuals at risk of developing COPD according to the concept of pre-COPD and compare their clinical characteristics with 1) those who have developed the disease at a young age, and 2) the overall population with and without COPD. Methods The EPISCAN II study is a cross-sectional, population-based study that aims to investigate the prevalence of COPD in Spain in subjects ≥40 years of age. Pre-COPD was defined as the presence of emphysema >5% and/or bronchial thickening by computed chromatography (CT) scan and/or diffusing capacity of the lung for carbon monoxide (D LCO) <80% of predicted in subjects with respiratory symptoms and post-bronchodilator forced expiratory volume in 1 s/forced vital capacity (FEV1/FVC) >0.70. Young COPD was defined as FEV1/FVC <0.70 in a subject ≤50 years of age. Demographic and clinical characteristics were compared among pre-COPD, young COPD and the overall population with and without COPD. Results Among the 1077 individuals with FEV1/FVC <0.70, 65 (6.0%) were ≤50 years of age. Among the 8015 individuals with FEV1/FVC >0.70, 350 underwent both D LCO testing and chest CT scanning. Of those, 78 (22.3%) subjects fulfilled the definition of pre-COPD. Subjects with pre-COPD were older, predominantly women, less frequently active or ex-smokers, with less frequent previous diagnosis of asthma but with higher symptomatic burden than those with young COPD. Conclusions 22.3% of the studied population was at risk of developing COPD, with similar symptomatic and structural changes to those with well-established disease without airflow obstruction. This COPD at-risk population is different from those that develop COPD at a young age.
Collapse
Affiliation(s)
- Borja G. Cosío
- Department of Medicine, University of Balearic Islands, Palma, Spain,CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,These authors contributed equally,Corresponding author: Borja G. Cosío ()
| | - Ciro Casanova
- Servicio de Neumología, Hospital Universitario Nuestra Señora de Candelaria, Tenerife, Spain,These authors contributed equally
| | - Juan José Soler-Cataluña
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,Servicio de Neumología, Hospital Arnau de Vilanova-Lliria, Valencia, Spain
| | - Joan B. Soriano
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,Servicio de Neumología, Hospital Universitario La Princesa and Universidad Autónoma de Madrid, Madrid, Spain
| | - Francisco García-Río
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,Servicio de Neumología, Hospital Universitario La Paz-IdiPAZ, Madrid, Spain
| | - Pilar de Lucas
- Servicio de Neumología, Hospital General Gregorio Marañon, Madrid, Spain
| | - Inmaculada Alfageme
- Unidad de Gestión Clínica de Neumología, Hospital Universitario Virgen de Valme, Universidad de Sevilla, Seville, Spain
| | | | | | - Julio Ancochea
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,Servicio de Neumología, Hospital Universitario La Princesa and Universidad Autónoma de Madrid, Madrid, Spain
| | - Marc Miravitlles
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain,Pneumology Department, Hospital Universitari Vall dHebron/Vall d'Hebron Institut de Recerca, Barcelona, Spain
| |
Collapse
|
33
|
He M, Zhang G, Shen F, Li X. Effects of Z-VaD-Ala-Asp-Fluoromethyl Ketone (Z-VAD-FMK) and Acetyl-Asp-Glu-Val-Asp-Aldehyde(Ac-DEVD-CHO) on Inflammation and Mucus Secretion in Mice Exposed to Cigarette Smoke. Int J Chron Obstruct Pulmon Dis 2023; 18:69-78. [PMID: 36777242 PMCID: PMC9910210 DOI: 10.2147/copd.s385748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/05/2023] [Indexed: 02/07/2023] Open
Abstract
Background and Objectives Smoking can lead to airway inflammation and mucus secretion through the nucleotide-binding domain-like receptor protein 3/caspase-1 pathway. In this study, z-VaD-Ala-Asp-fluoromethyl ketone(Z-VAD), a pan-caspase inhibitor, and acetyl-Asp-Glu-Val-Asp-aldehyde(Ac-DEVD), a caspase-3 inhibitor, were used to investigate the effect of caspase inhibitors on the expression of interleukin(IL)-1β and IL-8, airway inflammation, and mucus secretion in mice exposed to cigarette smoke(CS). Methods Thirty-two C57BL/6J male mice were divided into a control group, Smoke group, Z-VAD group, and Ac-DEVD group. Except for the control group, the animals were all exposed to CS for three months. After the experiment, lung function was measured and hematoxylin and eosin staining and periodic acid-Schiff staining were performed. The levels of IL-1β, IL-8, and mucin 5ac(Muc5ac) in serum and bronchoalveolar lavage fluid(BALF) were determined by enzyme-linked immunosorbent assay. Results Compared with the control group, the lung function of mice exposed to smoke was poorer, with a large number of inflammatory cells infiltrating around the airway, collapse of alveoli, expansion and fusion of distal alveoli, and formation of emphysema. The Z-VAD group was relieved compared with the smoke group. Airway inflammation was also reduced in the Ac-DEVD group compared with the Smoke group, but the degree of emphysema was not significantly improved. Although Z-VAD relieved airway inflammation and emphysema, Ac-DEVD only relieved inflammation. Z-VAD and Ac-DEVD decreased serum IL-1β and IL-8 levels. In BALF, IL-1β was decreased in Z-VAD group and IL-8 was highest in Smoke +Ac-DEVD group compared with control group and Ac-DEVD group. There was no significant difference in the expression of Muc5ac in serum. However, in BALF, levels of Muc5ac were higher in the smoking group and the lowest in the Ac-DEVD group. Conclusion Mice exposed to smoke had decreased lung function and significant cilia lodging, epithelial cell shedding, and inflammatory cell infiltration, with significant emphysema formation. The pan-caspase inhibitor, Z-VAD, improved airway inflammation and emphysema lesions in the mice exposed to smoke and reduced IL-1β and IL-8 levels in serum. The caspase-3 inhibitor, Ac-DEVD, reduced airway inflammation, serum IL-1β and IL-8 levels, and Muc5ac levels in BALF, but it did not improve emphysema.
Collapse
Affiliation(s)
- Mudan He
- Department of Respiratory, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, People’s Republic of China
| | - Guoqing Zhang
- Department of Respiratory, Jiading Branch of Shanghai General Hospital, Shanghai Jiao tong University School of Medicine, Shanghai, 200803, People’s Republic of China
| | - Fang Shen
- Department of Respiratory, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, People’s Republic of China
| | - Xingjing Li
- Department of Respiratory, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, People’s Republic of China,Correspondence: Xingjing Li, Department of Respiratory, Zhongshan Hospital Wusong Branch, Fudan University, No. 101 of North Tongtai Road, Baoshan District, Shanghai, 201900, People’s Republic of China, Tel +86 13816446543, Email
| |
Collapse
|
34
|
Agusti A, Ambrosino N, Blackstock F, Bourbeau J, Casaburi R, Celli B, Crouch R, Negro RD, Dreher M, Garvey C, Gerardi D, Goldstein R, Hanania N, Holland AE, Kaur A, Lareau S, Lindenauer PK, Mannino D, Make B, Maltais F, Marciniuk JD, Meek P, Morgan M, Pepin JL, Reardon JZ, Rochester C, Singh S, Spruit MA, Steiner MC, Troosters T, Vitacca M, Clini E, Jardim J, Nici L, Raskin J, ZuWallack R. COPD: Providing the right treatment for the right patient at the right time. Respir Med 2023; 207:107041. [PMID: 36610384 DOI: 10.1016/j.rmed.2022.107041] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a common disease associated with significant morbidity and mortality that is both preventable and treatable. However, a major challenge in recognizing, preventing, and treating COPD is understanding its complexity. While COPD has historically been characterized as a disease defined by airflow limitation, we now understand it as a multi-component disease with many clinical phenotypes, systemic manifestations, and associated co-morbidities. Evidence is rapidly emerging in our understanding of the many factors that contribute to the pathogenesis of COPD and the identification of "early" or "pre-COPD" which should provide exciting opportunities for early treatment and disease modification. In addition to breakthroughs in our understanding of the origins of COPD, we are optimizing treatment strategies and delivery of care that are showing impressive benefits in patient-centered outcomes and healthcare utilization. This special issue of Respiratory Medicine, "COPD: Providing the Right Treatment for the Right Patient at the Right Time" is a summary of the proceedings of a conference held in Stresa, Italy in April 2022 that brought together international experts to discuss emerging evidence in COPD and Pulmonary Rehabilitation in honor of a distinguished friend and colleague, Claudio Ferdinando Donor (1948-2021). Claudio was a true pioneer in the field of pulmonary rehabilitation and the comprehensive care of individuals with COPD. He held numerous leadership roles in in the field, provide editorial stewardship of several respiratory journals, authored numerous papers, statement and guidelines in COPD and Pulmonary Rehabilitation, and provided mentorship to many in our field. Claudio's most impressive talent was his ability to organize spectacular conferences and symposia that highlighted cutting edge science and clinical medicine. It is in this spirit that this conference was conceived and planned. These proceedings are divided into 4 sections which highlight crucial areas in the field of COPD: (1) New concepts in COPD pathogenesis; (2) Enhancing outcomes in COPD; (3) Non-pharmacologic management of COPD; and (4) Optimizing delivery of care for COPD. These presentations summarize the newest evidence in the field and capture lively discussion on the exciting future of treating this prevalent and impactful disease. We thank each of the authors for their participation and applaud their efforts toward pushing the envelope in our understanding of COPD and optimizing care for these patients. We believe that this edition is a most fitting tribute to a dear colleague and friend and will prove useful to students, clinicians, and researchers as they continually strive to provide the right treatment for the right patient at the right time. It has been our pleasure and a distinct honor to serve as editors and oversee such wonderful scholarly work.
Collapse
Affiliation(s)
- Alvar Agusti
- Clinic Barcelona Hospital University, Barcelona, Spain.
| | | | | | - Jean Bourbeau
- Department of Medicine, Division of Experimental Medicine, McGill University Health Centre, Montreal, QC, CA, USA.
| | | | | | | | - Roberto Dal Negro
- National Centre for Pharmacoeconomics and Pharmacoepidemiology (CESFAR), Verona, Italy.
| | - Michael Dreher
- Clinic of Cardiology, Angiology, Pneumology and Intensive Medicine, University Hospital Aachen, Aachen, 52074, DE, USA.
| | | | | | - Roger Goldstein
- Respiratory Rehabilitation Service, West Park Health Care Centre, Toronto, Ontario, CA, USA.
| | | | - Anne E Holland
- Departments of Physiotherapy and Respiratory Medicine, Alfred Health, Melbourne, Australia; Central Clinical School, Monash University, Melbourne, Australia; Institute for Breathing and Sleep, Melbourne, Australia.
| | - Antarpreet Kaur
- Section of Pulmonary, Critical Care, and Sleep Medicine, Trinity Health of New England, Hartford, CT, USA; University of Colorado School of Nursing, Aurora, CO, USA.
| | - Suzanne Lareau
- University of Colorado School of Nursing, Aurora, CO, USA.
| | - Peter K Lindenauer
- Department of Healthcare Delivery and Population Sciences, University of Massachusetts Chan Medical School - Baystate, Springfield, MA, USA.
| | | | - Barry Make
- National Jewish Health, Denver, CO, USA.
| | - François Maltais
- Institut Universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec, CA, USA.
| | - Jeffrey D Marciniuk
- Division of Respirology, Critical Care and Sleep Medicine, Department of Medicine, University of Saskatchewan, Saskatoon, CA, USA.
| | - Paula Meek
- University of Utah College of Nursing, Salt Lake City, UT, USA.
| | - Mike Morgan
- Dept of Respiratory Medicine, University Hospitals of Leicester, UK.
| | - Jean-Louis Pepin
- CHU de Grenoble - Clin Univ. de physiologie, sommeil et exercice, Grenoble, France.
| | - Jane Z Reardon
- Section of Pulmonary, Critical Care, and Sleep Medicine, Trinity Health of New England, Hartford, CT, USA.
| | | | - Sally Singh
- Department of Respiratory Diseases, University of Leicester, UK.
| | | | - Michael C Steiner
- Department of Respiratory Sciences, Leicester NIHR Biomedical Research Centre, Professor, University of Leicester, UK.
| | - Thierry Troosters
- Laboratory of Respiratory Diseases and Thoracic Surgery, KU Leuven: Leuven, Vlaanderen, Belgium.
| | - Michele Vitacca
- Department of Respiratory Rehabilitation, ICS S. Maugeri Care and Research Institutes, IRCCS Pavia, Italy.
| | - Enico Clini
- University of Modena and Reggio Emilia, Italy.
| | - Jose Jardim
- Federal University of Sao Paulo Paulista, Brazil.
| | - Linda Nici
- nBrown University School of Medicine, USA.
| | | | - Richard ZuWallack
- Section of Pulmonary, Critical Care, and Sleep Medicine, Saint Francis Hospital and Medical Center, 114 Woodland Street, Hartford, CT, 06105, USA.
| |
Collapse
|
35
|
The establishment of COPD organoids to study host-pathogen interaction reveals enhanced viral fitness of SARS-CoV-2 in bronchi. Nat Commun 2022; 13:7635. [PMID: 36496442 PMCID: PMC9735280 DOI: 10.1038/s41467-022-35253-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterised by airflow limitation and infective exacerbations, however, in-vitro model systems for the study of host-pathogen interaction at the individual level are lacking. Here, we describe the establishment of nasopharyngeal and bronchial organoids from healthy individuals and COPD that recapitulate disease at the individual level. In contrast to healthy organoids, goblet cell hyperplasia and reduced ciliary beat frequency were observed in COPD organoids, hallmark features of the disease. Single-cell transcriptomics uncovered evidence for altered cellular differentiation trajectories in COPD organoids. SARS-CoV-2 infection of COPD organoids revealed more productive replication in bronchi, the key site of infection in severe COVID-19. Viral and bacterial exposure of organoids induced greater pro-inflammatory responses in COPD organoids. In summary, we present an organoid model that recapitulates the in vivo physiological lung microenvironment at the individual level and is amenable to the study of host-pathogen interaction and emerging infectious disease.
Collapse
|
36
|
Thompson PJ, Criner GJ, Dransfield MT, Halpin DMG, Han MK, Lipson DA, Maghzal GJ, Martinez FJ, Midwinter D, Singh D, Tombs L, Wise RA. Effect of chronic mucus hypersecretion on treatment responses to inhaled therapies in patients with chronic obstructive pulmonary disease: Post hoc analysis of the IMPACT trial. Respirology 2022; 27:1034-1044. [PMID: 35970518 PMCID: PMC9804213 DOI: 10.1111/resp.14339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 07/18/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Chronic mucus hypersecretion (CMH) is a clinical phenotype of COPD. This exploratory post hoc analysis assessed relationship between CMH status and treatment response in IMPACT. METHODS Patients were randomized to once-daily fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) 100/62.5/25 μg, FF/VI 100/25 μg or UMEC/VI 62.5/25 μg and designated CMH+ if they scored 1/2 in St George's Respiratory Questionnaire (SGRQ) questions 1 and 2. Endpoints assessed by baseline CMH status included on-treatment exacerbation rates, change from baseline in trough forced expiratory volume in 1 second, SGRQ total score, COPD Assessment Test (CAT) score, proportion of SGRQ and CAT responders at Week 52 and safety. RESULTS Of 10,355 patients in the intent-to-treat population, 10,250 reported baseline SGRQ data (CMH+: 62% [n = 6383]). FF/UMEC/VI significantly (p < 0.001) reduced on-treatment moderate/severe exacerbation rates versus FF/VI and UMEC/VI in CMH+ (rate ratio: 0.87 and 0.72) and CMH- patients (0.82 and 0.80). FF/UMEC/VI significantly (p < 0.05) reduced on-treatment severe exacerbation rates versus UMEC/VI in CMH+ (0.62) and CMH- (0.74) subgroups. Similar improvements in health status and lung function with FF/UMEC/VI were observed, regardless of CMH status. In CMH+ patients, FF/VI significantly (p < 0.001) reduced on-treatment moderate/severe and severe exacerbation rates versus UMEC/VI (0.83 and 0.70). CONCLUSION FF/UMEC/VI had a favourable benefit: risk profile versus dual therapies irrespective of CMH status. The presence of CMH did not influence treatment response or exacerbations, lung function and/or health status. However, CMH did generate differences when dual therapies were compared and the impact of CMH should be considered in future trial design.
Collapse
Affiliation(s)
| | - Gerard J. Criner
- Lewis Katz School of Medicine at Temple UniversityPhiladelphiaPennsylvaniaUSA
| | - Mark T. Dransfield
- Division of Pulmonary, Allergy, and Critical Care Medicine, Lung Health CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - David M. G. Halpin
- University of Exeter Medical School, College of Medicine and HealthUniversity of ExeterExeterUK
| | - MeiLan K. Han
- Pulmonary & Critical CareUniversity of MichiganAnn ArborMichiganUSA
| | - David A. Lipson
- GlaxoSmithKlineCollegevillePennsylvaniaUSA,Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | | | | | - Dave Singh
- Centre for Respiratory Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science CentreThe University of Manchester, Manchester University NHS Foundation Hospital TrustManchesterUK
| | | | - Robert A. Wise
- Division of Pulmonary and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
37
|
Celli B, Fabbri L, Criner G, Martinez FJ, Mannino D, Vogelmeier C, Montes de Oca M, Papi A, Sin DD, Han MK, Agusti A. Definition and Nomenclature of Chronic Obstructive Pulmonary Disease: Time for Its Revision. Am J Respir Crit Care Med 2022; 206:1317-1325. [PMID: 35914087 PMCID: PMC9746870 DOI: 10.1164/rccm.202204-0671pp] [Citation(s) in RCA: 234] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/01/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Bartolome Celli
- Pulmonary Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Leonardo Fabbri
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Gerard Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Fernando J Martinez
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David Mannino
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Kentucky College of Medicine, Lexington, Kentucky
| | - Claus Vogelmeier
- Pulmonary and Critical Care Medicine, Department of Medicine, University Medical Center University of Marburg, German Center for Lung Research (DZL), Philipps University Marburg, Marburg, Germany
| | - Maria Montes de Oca
- Hospital Universitario de Caracas, Universidad Central de Venezuela and Centro Médico de Caracas, Caracas, Venezuela
| | - Alberto Papi
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Don D Sin
- Division of Respiratory Medicine, Centre for Heart Lung Innovation, St. Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - MeiLan K Han
- University of Michigan Health System, Ann Arbor, Michigan; and
| | - Alvar Agusti
- Cátedra Salud Respiratoria, Universitat de Barcelona; Respiratory Institute, Hospital Clinic, Barcelona; IDIBAPS, CIBERES, Barcelona, Spain
| |
Collapse
|
38
|
Celli BR, Singh D, Vogelmeier C, Agusti A. New Perspectives on Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2022; 17:2127-2136. [PMID: 36097591 PMCID: PMC9464005 DOI: 10.2147/copd.s365771] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide; many recent advances have been made in many aspects of the disease. The aim of this article is to illustrate and discuss some of these advances in the management of different types of patients. Large-scale trials have confirmed that long-acting bronchodilator therapy, particularly using the combination of LABA/LAMA, remains the mainstay of COPD treatment, with special attention being paid to careful selection of inhaler devices. The initial choice of pharmacological therapy is based on the GOLD ABCD grouping of patients. It is very important to stress that there is a need to implement a management cycle because COPD is a chronic disease with varying clinical course and a high number of potential comorbidities that may affect morbidity and mortality. Therefore, regular reevaluation of the patient is mandatory. This allows identification of characteristics aimed at maximizing the benefits for a specific patient or a subset of patients. Within this context, the role of the blood eosinophil count as a marker of inhaled corticosteroids response to prevent future exacerbations in patients who, despite appropriate bronchodilator therapy, still suffer from them has been proven to be a useful simple biomarker in medication selection. These advances support the concept of precision medicine, with the goal that patients get the right medicine at the right time for the right reason. Finally, recent studies have shown that early life events may be of critical relevance for the development of COPD. With this as a background, concepts to identify individuals at risk and early identification of cases have become an important objective of current research with the hope of maximizing the effects of therapy and the possibility of impacting disease progression.
Collapse
Affiliation(s)
| | - Dave Singh
- Division of Infection, Immunity & Respiratory Medicine, University of Manchester, Manchester University NHS Hospital Trust, Manchester, UK
| | - Claus Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, German Center for Lung Research (DZL), University of Marburg, Marburg, Germany
| | - Alvar Agusti
- Respiratory Institute, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERES, Barcelona, Spain
| |
Collapse
|
39
|
Casara A, Turato G, Marin-Oto M, Semenzato U, Biondini D, Tinè M, Bernardinello N, Cocconcelli E, Cubero P, Balestro E, Spagnolo P, Marin JM, Cosio MG, Saetta M, Bazzan E. Chronic Bronchitis Affects Outcomes in Smokers without Chronic Obstructive Pulmonary Disease (COPD). J Clin Med 2022; 11:jcm11164886. [PMID: 36013126 PMCID: PMC9410001 DOI: 10.3390/jcm11164886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
Background. Chronic bronchitis (CB) importantly affects outcomes in smokers with COPD, but the effects on smokers without COPD are less well known and less emphasized. The aim of our study was to investigate the possible effects of CB on clinical outcomes in smokers without COPD (noCOPD) and compare them with the effects in smokers with COPD (COPD). Methods. For that purpose, we studied 511 smokers, 302 with and 209 without COPD, followed for 10 years in an academic COPD ambulatory setting. Chronic bronchitis was defined as the presence of cough and sputum production for at least 3 months in each of two consecutive years. All subjects underwent clinical and functional examination with spirometry, diffusion capacity (DLco), 6-min walking test (6MWT), mMRC Dyspnoea Scale, COPD Assessment Test (CAT), and recording of annual frequency of exacerbations. All-cause mortality during follow-up was recorded. Results. 27% of noCOPD and 45% of COPD had CB. noCOPD with CB had lower FEV1 and DLco, worse 6MWT, more dyspnoea, a higher number of exacerbations and lower survival than noCOPD without CB. CB did not affect FEV1 decline in noCOPD but it significantly did in COPD. Conclusions. The presence of chronic bronchitis in smokers without COPD will significantly affect symptoms, quality of life, and survival, underlining the importance of recognizing the condition and managing it accordingly.
Collapse
Affiliation(s)
- Alvise Casara
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Graziella Turato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Marta Marin-Oto
- Respiratory Service, Hospital Clinico Universitario, 50009 Zaragoza, Spain
| | - Umberto Semenzato
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Davide Biondini
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Mariaenrica Tinè
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Nicol Bernardinello
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Elisabetta Cocconcelli
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Pablo Cubero
- Translational Research Unit (IIS Aragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
| | - Elisabetta Balestro
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Paolo Spagnolo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Josè M. Marin
- Translational Research Unit (IIS Aragón), Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain
- Department of Medicine, University of Zaragoza School of Medicine, 50009 Zaragoza, Spain
| | - Manuel G. Cosio
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Meakins-Christie Laboratories, Respiratory Division, McGill University, Montreal, QU 000004, Canada
| | - Marina Saetta
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Erica Bazzan
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
- Correspondence:
| |
Collapse
|
40
|
Respiratory Symptoms among US Adults: a Cross-Sectional Health Survey Study. Pulm Ther 2022; 8:255-268. [PMID: 35794458 PMCID: PMC9458821 DOI: 10.1007/s41030-022-00194-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/23/2022] [Indexed: 10/26/2022] Open
Abstract
INTRODUCTION Data collected through ongoing, state-based, cross-sectional health surveys could be used to better understand the contribution of respiratory symptoms to impaired health among the US adult population. METHODS We used the 2015 Behavioral Risk Factor Surveillance System telephone health survey in four states (Kentucky, Florida, South Carolina, Texas) to describe the relationship between symptoms, associated factors such as tobacco smoking, and health impairments. Self-reported productive cough, shortness of breath (SOB), and dyspnea on exertion (DOE) were categorized as minimal, moderate, or severe. Data were analyzed using multiple logistic regression models with age as a covariate to assess relationships of symptoms with other factors. RESULTS Among adults ≥ 18 years, respiratory impairment [current asthma, chronic obstructive pulmonary disease (COPD), or a current moderate or severe symptom] occurred in 39.1% of the population. More than half of adults reporting moderate or severe symptoms had not been diagnosed with asthma or COPD, particularly with DOE and productive cough. Subjects were at greater risk of moderate and severe SOB or productive cough with increasing age, prolonged smoking duration (≥ 20 years), being an ever-smoker, or if reporting COPD, current asthma, or any other comorbidity except cancer. Morbid obesity [body mass index (BMI) > 35 kg/m2] was associated with severe DOE at a rate similar to current asthma or COPD (25.6%, 95% CI 20.9-30.3%; 20.8%, 95% CI 16.4-25.1%; 21.3%, 95% CI 17.5-25.1%, respectively); it was the most common cause of DOE. SOB was associated with worse general health impairment and limited ambulation compared with other symptoms. Tobacco smoking prevalence and race varied among states, affecting symptom prevalence. CONCLUSION In the largest US survey in decades, we provide a current perspective of respiratory symptoms among adults of all ages. While known risk factors were apparent, low-risk persons also frequently reported symptoms and impairments.
Collapse
|
41
|
Guerra S. Nonobstructive Chronic Bronchitis. Chest 2022; 162:19-21. [DOI: 10.1016/j.chest.2022.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/28/2022] Open
|
42
|
Calzetta L, Ritondo BL, Zappa MC, Manzetti GM, Perduno A, Shute J, Rogliani P. The impact of long-acting muscarinic antagonists on mucus hypersecretion and cough in chronic obstructive pulmonary disease: a systematic review. Eur Respir Rev 2022; 31:31/164/210196. [PMID: 35508331 DOI: 10.1183/16000617.0196-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/20/2022] [Indexed: 01/18/2023] Open
Abstract
Patients suffering from chronic obstructive pulmonary disease (COPD) clinically manifest airway mucus hypersecretion as sputum expectoration and cough. Evidence accumulated in the past decade has shown that the cholinergic system not only regulates airway smooth muscle contraction but also the activity of inflammatory and airway epithelial cells, including goblet cells, and submucosal gland activity. Long-acting muscarinic antagonists (LAMAs) with the most favourable M3/M2 muscarinic acetylcholine (ACh) receptors residency properties are not only excellent bronchodilators but potentially also mucus-modifying agents, able to positively impact on mucus hypersecretion and cough. The aim of this systematic review was to investigate the impact of LAMAs on mucus hypersecretion and cough in COPD patients. The evidence confirmed that LAMAs, mainly tiotropium and aclidinium, improved sputum production and cough in moderate to severe COPD. Thus, LAMAs not only antagonise the ACh-induced bronchoconstriction of the airways but also appear to limit the production of mucus secreted in response to ACh by airway goblet cells and/or submucosal glands. Further clinical studies are necessary to evaluate the impact of LAMAs exclusively on sputum symptoms and cough as primary end-points and to investigate whether LAMAs have a modulatory action on the rheological properties of mucus.
Collapse
Affiliation(s)
- Luigino Calzetta
- Dept of Medicine and Surgery, Respiratory Disease and Lung Function Unit, University of Parma, Parma, Italy
| | - Beatrice Ludovica Ritondo
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Gian Marco Manzetti
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Andrea Perduno
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Janis Shute
- School of Pharmacy and Biomedical Science, University of Portsmouth, Portsmouth, UK
| | - Paola Rogliani
- Unit of Respiratory Medicine, Dept of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
43
|
Christenson SA, Smith BM, Bafadhel M, Putcha N. Chronic obstructive pulmonary disease. Lancet 2022; 399:2227-2242. [PMID: 35533707 DOI: 10.1016/s0140-6736(22)00470-6] [Citation(s) in RCA: 503] [Impact Index Per Article: 167.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 02/16/2022] [Accepted: 02/25/2022] [Indexed: 12/14/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity, mortality, and health-care use worldwide. COPD is caused by exposure to inhaled noxious particles, notably tobacco smoke and pollutants. However, the broad range of factors that increase the risk of development and progression of COPD throughout the life course are increasingly being recognised. Innovations in omics and imaging techniques have provided greater insight into disease pathobiology, which might result in advances in COPD prevention, diagnosis, and treatment. Although few novel treatments have been approved for COPD in the past 5 years, advances have been made in targeting existing therapies to specific subpopulations using new biomarker-based strategies. Additionally, COVID-19 has undeniably affected individuals with COPD, who are not only at higher risk for severe disease manifestations than healthy individuals but also negatively affected by interruptions in health-care delivery and social isolation. This Seminar reviews COPD with an emphasis on recent advances in epidemiology, pathophysiology, imaging, diagnosis, and treatment.
Collapse
Affiliation(s)
- Stephanie A Christenson
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Benjamin M Smith
- Department of Medicine, Columbia University Medical Center, New York, NY, USA; Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Mona Bafadhel
- School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK; Department of Respiratory Medicine, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Perret JL, Wurzel D, Walters EH, Lowe AJ, Lodge CJ, Bui DS, Erbas B, Bowatte G, Russell MA, Thompson BR, Gurrin L, Thomas PS, Hamilton G, Hopper JL, Abramson MJ, Chang AB, Dharmage SC. Childhood 'bronchitis' and respiratory outcomes in middle-age: a prospective cohort study from age 7 to 53 years. BMJ Open Respir Res 2022; 9:e001212. [PMID: 35725733 PMCID: PMC9240942 DOI: 10.1136/bmjresp-2022-001212] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/14/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Chronic bronchitis in childhood is associated with a diagnosis of asthma and/or bronchiectasis a few years later, however, consequences into middle-age are unknown. OBJECTIVE To investigate the relationship between childhood bronchitis and respiratory-related health outcomes in middle-age. DESIGN Cohort study from age 7 to 53 years. SETTING General population of European descent from Tasmania, Australia. PARTICIPANTS 3202 participants of the age 53-year follow-up (mean age 53, range 51-55) of the Tasmanian Longitudinal Health Study cohort who were born in 1961 and first investigated at age 7 were included in our analysis. STATISTICAL METHODS Multivariable linear and logistic regression. The association between parent reported childhood bronchitis up to age 7 and age 53-year lung conditions (n=3202) and lung function (n=2379) were investigated. RESULTS Among 3202 participants, 47.5% had one or more episodes of childhood bronchitis, classified according to severity based on the number of episodes and duration as: 'non-recurrent bronchitis' (28.1%); 'recurrent non-protracted bronchitis' (18.1%) and 'recurrent-protracted bronchitis' (1.3%). Age 53 prevalence of doctor-diagnosed asthma and pneumonia (p-trend <0.001) and chronic bronchitis (p-trend=0.07) increased in accordance with childhood bronchitis severities. At age 53, 'recurrent-protracted bronchitis' (the most severe subgroup in childhood) was associated with doctor-diagnosed current asthma (OR 4.54, 95% CI 2.31 to 8.91) doctor-diagnosed pneumonia (OR=2.18 (95% CI 1.00 to 4.74)) and, paradoxically, increased transfer factor for carbon monoxide (z-score +0.51 SD (0.15-0.88)), when compared with no childhood bronchitis. CONCLUSION In this cohort born in 1961, one or more episodes of childhood bronchitis was a frequent occurrence. 'Recurrent-protracted bronchitis', while uncommon, was especially linked to multiple respiratory outcomes almost five decades later, including asthma, pneumonia and raised lung gas transfer. These findings provide insights into the natural history of childhood 'bronchitis' into middle-age.
Collapse
Affiliation(s)
- Jennifer L Perret
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Respiratory and Sleep Medicine, The Austin Hospital, Melbourne, Victoria, Australia
- Institute for Breathing and Sleep (IBAS), Melbourne, Victoria, Australia
| | - Danielle Wurzel
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Respiratory Medicine, The Royal Children's Hospital, Melbourne, Victoria, Australia
- Infection and Immunity, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - E Haydn Walters
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Adrian J Lowe
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Caroline J Lodge
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dinh S Bui
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Bircan Erbas
- Department of Public Health, School of Psychology and Public Health, La Trobe University, Bundoora, Victoria, Australia
| | - Gayan Bowatte
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Melissa A Russell
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Bruce R Thompson
- School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Lyle Gurrin
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul S Thomas
- Prince of Wales' Clinical School, and Mechanisms of Disease and Translational Research, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Garun Hamilton
- Monash Lung, Sleep, Allergy and Immunology, Monash Health, Clayton, Victoria, Australia
- School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Anne B Chang
- Department of Respiratory Medicine, Queensland Children's Hospital, Brisbane, Queensland, Australia
- Child Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Agustí A, Melén E, DeMeo DL, Breyer-Kohansal R, Faner R. Pathogenesis of chronic obstructive pulmonary disease: understanding the contributions of gene-environment interactions across the lifespan. THE LANCET. RESPIRATORY MEDICINE 2022; 10:512-524. [PMID: 35427533 PMCID: PMC11428195 DOI: 10.1016/s2213-2600(21)00555-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 11/08/2021] [Accepted: 12/06/2021] [Indexed: 12/31/2022]
Abstract
The traditional view of chronic obstructive pulmonary disease (COPD) as a self-inflicted disease caused by tobacco smoking in genetically susceptible individuals has been challenged by recent research findings. COPD can instead be understood as the potential end result of the accumulation of gene-environment interactions encountered by an individual over the life course. Integration of a time axis in pathogenic models of COPD is necessary because the biological responses to and clinical consequences of different exposures might vary according to both the age of an individual at which a given gene-environment interaction occurs and the cumulative history of previous gene-environment interactions. Future research should aim to understand the effects of dynamic interactions between genes (G) and the environment (E) by integrating information from basic omics (eg, genomics, epigenomics, proteomics) and clinical omics (eg, phenomics, physiomics, radiomics) with exposures (the exposome) over time (T)-an approach that we refer to as GETomics. In the context of this approach, we argue that COPD should be viewed not as a single disease, but as a clinical syndrome characterised by a recognisable pattern of chronic symptoms and structural or functional impairments due to gene-environment interactions across the lifespan that influence normal lung development and ageing.
Collapse
Affiliation(s)
- Alvar Agustí
- Càtedra Salut Respiratòria, Universitat Barcelona, Barcelona, Spain; Respiratory Institute, Hospital Clinic, Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Dawn L DeMeo
- Channing Division of Network Medicine, and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Robab Breyer-Kohansal
- Ludwig Boltzmann Institute for Lung Health, Vienna, Austria; Department of Respiratory and Critical Care Medicine, Clinic Penzing, Vienna, Austria
| | - Rosa Faner
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Barcelona, Spain.
| |
Collapse
|
46
|
Radovanovic D, Contoli M, Braido F, Maniscalco M, Micheletto C, Solidoro P, Santus P, Carone M. Future Perspectives of Revaluating Mild COPD. Respiration 2022; 101:688-696. [PMID: 35468602 DOI: 10.1159/000524102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 03/07/2022] [Indexed: 11/19/2022] Open
Abstract
In 2020, COPD was the third leading cause of death worldwide. Lung function is central for the diagnosis of this disease, and COPD severity is still partially classified based on airflow obstruction, which can range from "mild" (GOLD 1 group, FEV1 ≥80% predicted) to "very severe" (GOLD 4, FEV1 <30% predicted). However, the term "mild COPD" needs to be carefully analyzed. Several studies have shown that even in the presence of a mild obstruction, patients can have significant symptoms, physiological deterioration, evidence of emphysema, and suffer from recurrent exacerbations. Small airways pathology significantly correlates with the presence of symptoms, and it has been demonstrated that the onset of bronchiolitis occurs earlier than that of emphysema. These damages have long been known to not be detectable with conventional tests, and exclusive reliance on spirometry is not enough to adequately study and stage a patient with "mild COPD." Therefore, early identification of COPD is of utmost importance in the light of modifying the natural course of the disease. However, patients with early lung damage are yet to be included and studied in interventional clinical trials.
Collapse
Affiliation(s)
- Dejan Radovanovic
- Division of Respiratory Diseases, Ospedale L. Sacco, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Marco Contoli
- Respiratory Diseases, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Fulvio Braido
- Respiratory Unit for Continuity of Care, IRCCS, Ospedale Policlinico San Martino, Department of Internal Medicine (DiMI), University of Genova, Genova, Italy
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri, Pavia. IRCCS di Telese, Telese Terme, Italy
| | - Claudio Micheletto
- UOC di Pneumologia, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Paolo Solidoro
- Medical Sciences Department, University of Torino, Turin, Italy.,Division of Respiratory Diseases, Cardiovascular and Thoracic Department, AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Pierachille Santus
- Division of Respiratory Diseases, Ospedale L. Sacco, ASST Fatebenefratelli-Sacco, Milan, Italy.,Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Mauro Carone
- Istituti Clinici Scientifici Maugeri, Pavia. IRCCS di Bari, Bari, Italy
| |
Collapse
|
47
|
Singanayagam A, Footitt J, Marczynski M, Radicioni G, Cross MT, Finney LJ, Trujillo-Torralbo MB, Calderazzo M, Zhu J, Aniscenko J, Clarke TB, Molyneaux PL, Bartlett NW, Moffatt MF, Cookson WO, Wedzicha J, Evans CM, Boucher RC, Kesimer M, Lieleg O, Mallia P, Johnston SL. Airway mucins promote immunopathology in virus-exacerbated chronic obstructive pulmonary disease. J Clin Invest 2022; 132:e120901. [PMID: 35239513 PMCID: PMC9012283 DOI: 10.1172/jci120901] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
The respiratory tract surface is protected from inhaled pathogens by a secreted layer of mucus rich in mucin glycoproteins. Abnormal mucus accumulation is a cardinal feature of chronic respiratory diseases, but the relationship between mucus and pathogens during exacerbations is poorly understood. We identified elevations in airway mucin 5AC (MUC5AC) and MUC5B concentrations during spontaneous and experimentally induced chronic obstructive pulmonary disease (COPD) exacerbations. MUC5AC was more sensitive to changes in expression during exacerbation and was therefore more predictably associated with viral load, inflammation, symptom severity, decrements in lung function, and secondary bacterial infections. MUC5AC was functionally related to inflammation, as Muc5ac-deficient (Muc5ac-/-) mice had attenuated RV-induced (RV-induced) airway inflammation, and exogenous MUC5AC glycoprotein administration augmented inflammatory responses and increased the release of extracellular adenosine triphosphate (ATP) in mice and human airway epithelial cell cultures. Hydrolysis of ATP suppressed MUC5AC augmentation of RV-induced inflammation in mice. Therapeutic suppression of mucin production using an EGFR antagonist ameliorated immunopathology in a mouse COPD exacerbation model. The coordinated virus induction of MUC5AC and MUC5B expression suggests that non-Th2 mechanisms trigger mucin hypersecretion during exacerbations. Our data identified a proinflammatory role for MUC5AC during viral infection and suggest that MUC5AC inhibition may ameliorate COPD exacerbations.
Collapse
Affiliation(s)
- Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Joseph Footitt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Matthias Marczynski
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Giorgia Radicioni
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michael T. Cross
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lydia J. Finney
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Maria Calderazzo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Julia Aniscenko
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas B. Clarke
- Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Philip L. Molyneaux
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nathan W. Bartlett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
- College of Health, Medicine and Wellbeing, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Miriam F. Moffatt
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - William O. Cookson
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jadwiga Wedzicha
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Christopher M. Evans
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Richard C. Boucher
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Marsico Lung Institute/Cystic Fibrosis and Pulmonary Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Oliver Lieleg
- School of Engineering and Design, Department of Materials Engineering and
- Center for Protein Assemblies, Technical University of Munich, Munich, Germany
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | |
Collapse
|
48
|
Çolak Y, Nordestgaard BG, Vestbo J, Afzal S, Lange P. Potential clinical implications of targeted spirometry for detection of COPD: A contemporary population-based cohort study. Respir Med 2022; 197:106852. [DOI: 10.1016/j.rmed.2022.106852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
|
49
|
Tajiri T, Matsumoto H, Jinnai M, Kanemitsu Y, Nagasaki T, Iwata T, Inoue H, Nakaji H, Oguma T, Ito I, Niimi A. Pathophysiological relevance of sputum MUC5AC and MUC5B levels in patients with mild asthma. Allergol Int 2022; 71:193-199. [PMID: 34656442 DOI: 10.1016/j.alit.2021.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/28/2021] [Accepted: 09/07/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Airway mucus hypersecretion is an important pathophysiological feature of asthma. MUC5AC and MUC5B are the major secreted polymeric mucins in airways, and their compositions affect mucus properties. Despite the increasing appreciation of MUC5AC and MUC5B compositions in asthmatic airways, their pathophysiological relevance remains to be fully understood in humans. METHODS In this cross-sectional study, we prospectively enrolled newly referred steroid-untreated patients with mild asthma and healthy controls. We compared induced sputum MUC5AC and MUC5B levels between patients and controls. Subsequently, we assessed the correlation between MUC5AC and MUC5B levels and clinical indices in patients. Sputum MUC5AC and MUC5B levels were measured using enzyme-linked immunosorbent assays. RESULTS Sputum MUC5AC and MUC5B levels were significantly higher in patients (n = 87) than in controls (n = 22) (p = 0.0002 and p = 0.006, respectively). The ratio of sputum MUC5AC to MUC5B tended to be higher in patients than in controls (p = 0.07). Sputum MUC5AC levels significantly and positively correlated with fractional exhaled nitric oxide at expiratory flow of 50 mL/s (Spearman's rho = 0.29, p = 0.006), sputum eosinophil proportion (rho = 0.34, p = 0.0013), and airway sensitivity (rho = 0.39, p = 0.0005). By contrast, sputum MUC5B levels significantly and positively correlated with airway sensitivity (rho = 0.35, p = 0.002) and negatively correlated with airway reactivity (rho = -0.33, p = 0.004). CONCLUSIONS Sputum MUC5AC is increased by protein levels and involved in airway type 2/eosinophilic inflammation and airway hyperresponsiveness in steroid-untreated patients with mild asthma.
Collapse
Affiliation(s)
- Tomoko Tajiri
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University School of Medical Sciences, Aichi, Japan.
| | - Hisako Matsumoto
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine and Allergology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Makiko Jinnai
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Kanemitsu
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University School of Medical Sciences, Aichi, Japan
| | - Tadao Nagasaki
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Toshiyuki Iwata
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Inoue
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Hitoshi Nakaji
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Isao Ito
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan
| | - Akio Niimi
- Department of Respiratory Medicine, Kyoto University, Kyoto, Japan; Department of Respiratory Medicine, Allergy and Clinical Immunology, Nagoya City University School of Medical Sciences, Aichi, Japan
| |
Collapse
|
50
|
Martinez FJ, Agusti A, Celli BR, Han MK, Allinson JP, Bhatt SP, Calverley P, Chotirmall SH, Chowdhury B, Darken P, Da Silva CA, Donaldson G, Dorinsky P, Dransfield M, Faner R, Halpin DM, Jones P, Krishnan JA, Locantore N, Martinez FD, Mullerova H, Price D, Rabe KF, Reisner C, Singh D, Vestbo J, Vogelmeier CF, Wise RA, Tal-Singer R, Wedzicha JA. Treatment Trials in Young Patients with Chronic Obstructive Pulmonary Disease and Pre-Chronic Obstructive Pulmonary Disease Patients: Time to Move Forward. Am J Respir Crit Care Med 2022; 205:275-287. [PMID: 34672872 PMCID: PMC8886994 DOI: 10.1164/rccm.202107-1663so] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 10/19/2021] [Indexed: 02/03/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the end result of a series of dynamic and cumulative gene-environment interactions over a lifetime. The evolving understanding of COPD biology provides novel opportunities for prevention, early diagnosis, and intervention. To advance these concepts, we propose therapeutic trials in two major groups of subjects: "young" individuals with COPD and those with pre-COPD. Given that lungs grow to about 20 years of age and begin to age at approximately 50 years, we consider "young" patients with COPD those patients in the age range of 20-50 years. Pre-COPD relates to individuals of any age who have respiratory symptoms with or without structural and/or functional abnormalities, in the absence of airflow limitation, and who may develop persistent airflow limitation over time. We exclude from the current discussion infants and adolescents because of their unique physiological context and COPD in older adults given their representation in prior randomized controlled trials (RCTs). We highlight the need of RCTs focused on COPD in young patients or pre-COPD to reduce disease progression, providing innovative approaches to identifying and engaging potential study subjects. We detail approaches to RCT design, including potential outcomes such as lung function, patient-reported outcomes, exacerbations, lung imaging, mortality, and composite endpoints. We critically review study design components such as statistical powering and analysis, duration of study treatment, and formats to trial structure, including platform, basket, and umbrella trials. We provide a call to action for treatment RCTs in 1) young adults with COPD and 2) those with pre-COPD at any age.
Collapse
Affiliation(s)
| | - Alvar Agusti
- Catedra Salut Respiratoria and
- Institut Respiratorio, Hospital Clinic, Barcelona, Spain
- Institut d’investigacions biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Bartolome R. Celli
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - MeiLan K. Han
- University of Michigan Health System, Ann Arbor, Michigan
| | - James P. Allinson
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Surya P. Bhatt
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Peter Calverley
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | | | | | | | - Carla A. Da Silva
- Clinical Development, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Gavin Donaldson
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | | | - Mark Dransfield
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rosa Faner
- Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | | | - Paul Jones
- St. George’s University of London, London, United Kingdom
| | | | | | | | | | - David Price
- Observational and Pragmatic Research Institute, Singapore
- Centre of Academic Primary Care, Division of Applied Health Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Klaus F. Rabe
- LungenClinic Grosshansdorf, Member of the German Center for Lung Research, Grosshansdorf, Germany
- Department of Medicine, Christian Albrechts University Kiel, Member of the German Center for Lung Research Kiel, Germany
| | | | | | - Jørgen Vestbo
- Manchester University NHS Trust, Manchester, United Kingdom
| | - Claus F. Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University of Marburg, Member of the German Center for Lung Research, Marburg, Germany
| | | | | | | |
Collapse
|