301
|
Begum F, Ruczinski I, Li S, Silverman EK, Cho MH, Lynch DA, Curran-Everett D, Crapo J, Scharpf RB, Parker MM, Hetmanski JB, Beaty TH. Identifying a Deletion Affecting Total Lung Capacity Among Subjects in the COPDGene Study Cohort. Genet Epidemiol 2015; 40:81-8. [PMID: 26643968 DOI: 10.1002/gepi.21943] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 09/21/2015] [Accepted: 10/19/2015] [Indexed: 01/17/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive disease with both environmental and genetic risk factors. Genome-wide association studies (GWAS) have identified multiple genomic regions influencing risk of COPD. To thoroughly investigate the genetic etiology of COPD, however, it is also important to explore the role of copy number variants (CNVs) because the presence of structural variants can alter gene expression and can be causal for some diseases. Here, we investigated effects of polymorphic CNVs on quantitative measures of pulmonary function and chest computed tomography (CT) phenotypes among subjects enrolled in COPDGene, a multisite study. COPDGene subjects consist of roughly one-third African American (AA) and two-thirds non-Hispanic white adult smokers (with or without COPD). We estimated CNVs using PennCNV on 9,076 COPDGene subjects using Illumina's Omni-Express genome-wide marker array. We tested for association between polymorphic CNV components (defined as disjoint intervals of copy number regions) for several quantitative phenotypes associated with COPD within each racial group. Among the AAs, we identified a polymorphic CNV on chromosome 5q35.2 located between two genes (FAM153B and SIMK1, but also harboring several pseudo-genes) giving genome-wide significance in tests of association with total lung capacity (TLCCT ) as measured by chest CT scans. This is the first study of genome-wide association tests of polymorphic CNVs and TLCCT . Although the ARIC cohort did not have the phenotype of TLCCT , we found similar counts of CNV deletions and amplifications among AA and European subjects in this second cohort.
Collapse
Affiliation(s)
- Ferdouse Begum
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Shengchao Li
- Cancer Genomics Research Laboratory (CGR), Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland, United States of America
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - David A Lynch
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Douglas Curran-Everett
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, United States of America
| | - James Crapo
- Department of Medicine, National Jewish Health, Denver, Colorado, United States of America
| | - Robert B Scharpf
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Margaret M Parker
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jacqueline B Hetmanski
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| |
Collapse
|
302
|
Lutz SM, Cho MH, Young K, Hersh CP, Castaldi PJ, McDonald ML, Regan E, Mattheisen M, DeMeo DL, Parker M, Foreman M, Make BJ, Jensen RL, Casaburi R, Lomas DA, Bhatt SP, Bakke P, Gulsvik A, Crapo JD, Beaty TH, Laird NM, Lange C, Hokanson JE, Silverman EK. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet 2015; 16:138. [PMID: 26634245 PMCID: PMC4668640 DOI: 10.1186/s12863-015-0299-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/20/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pulmonary function decline is a major contributor to morbidity and mortality among smokers. Post bronchodilator FEV1 and FEV1/FVC ratio are considered the standard assessment of airflow obstruction. We performed a genome-wide association study (GWAS) in 9919 current and former smokers in the COPDGene study (6659 non-Hispanic Whites [NHW] and 3260 African Americans [AA]) to identify associations with spirometric measures (post-bronchodilator FEV1 and FEV1/FVC). We also conducted meta-analysis of FEV1 and FEV1/FVC GWAS in the COPDGene, ECLIPSE, and GenKOLS cohorts (total n = 13,532). RESULTS Among NHW in the COPDGene cohort, both measures of pulmonary function were significantly associated with SNPs at the 15q25 locus [containing CHRNA3/5, AGPHD1, IREB2, CHRNB4] (lowest p-value = 2.17 × 10(-11)), and FEV1/FVC was associated with a genomic region on chromosome 4 [upstream of HHIP] (lowest p-value = 5.94 × 10(-10)); both regions have been previously associated with COPD. For the meta-analysis, in addition to confirming associations to the regions near CHRNA3/5 and HHIP, genome-wide significant associations were identified for FEV1 on chromosome 1 [TGFB2] (p-value = 8.99 × 10(-9)), 9 [DBH] (p-value = 9.69 × 10(-9)) and 19 [CYP2A6/7] (p-value = 3.49 × 10(-8)) and for FEV1/FVC on chromosome 1 [TGFB2] (p-value = 8.99 × 10(-9)), 4 [FAM13A] (p-value = 3.88 × 10(-12)), 11 [MMP3/12] (p-value = 3.29 × 10(-10)) and 14 [RIN3] (p-value = 5.64 × 10(-9)). CONCLUSIONS In a large genome-wide association study of lung function in smokers, we found genome-wide significant associations at several previously described loci with lung function or COPD. We additionally identified a novel genome-wide significant locus with FEV1 on chromosome 9 [DBH] in a meta-analysis of three study populations.
Collapse
Affiliation(s)
- Sharon M Lutz
- Department of Biostatistics, University of Colorado Anschutz Medical Campus, 13001 E. 17th Place, B119 Bldg. 500, W3128, Aurora, CO, 80045, USA.
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Kendra Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Merry-Lynn McDonald
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Elizabeth Regan
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| | - Manuel Mattheisen
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Margaret Parker
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | | | - Barry J Make
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| | - Robert L Jensen
- Division of Pulmonary, Allergy & Critical Care Medicine, LDS Hospital, Salt Lake City, UT, USA.
| | - Richard Casaburi
- Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA.
| | - David A Lomas
- Wolfson Institute for Biomedical Research, University College London, London, UK.
| | - Surya P Bhatt
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| | - Terri H Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Nan M Laird
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA.
| | - Christoph Lange
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA.
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
303
|
Identification of five chronic obstructive pulmonary disease subgroups with different prognoses in the ECLIPSE cohort using cluster analysis. Ann Am Thorac Soc 2015; 12:303-12. [PMID: 25642832 DOI: 10.1513/annalsats.201403-125oc] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease that likely includes clinically relevant subgroups. OBJECTIVES To identify subgroups of COPD in ECLIPSE (Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints) subjects using cluster analysis and to assess clinically meaningful outcomes of the clusters during 3 years of longitudinal follow-up. METHODS Factor analysis was used to reduce 41 variables determined at recruitment in 2,164 patients with COPD to 13 main factors, and the variables with the highest loading were used for cluster analysis. Clusters were evaluated for their relationship with clinically meaningful outcomes during 3 years of follow-up. The relationships among clinical parameters were evaluated within clusters. MEASUREMENTS AND MAIN RESULTS Five subgroups were distinguished using cross-sectional clinical features. These groups differed regarding outcomes. Cluster A included patients with milder disease and had fewer deaths and hospitalizations. Cluster B had less systemic inflammation at baseline but had notable changes in health status and emphysema extent. Cluster C had many comorbidities, evidence of systemic inflammation, and the highest mortality. Cluster D had low FEV1, severe emphysema, and the highest exacerbation and COPD hospitalization rate. Cluster E was intermediate for most variables and may represent a mixed group that includes further clusters. The relationships among clinical variables within clusters differed from that in the entire COPD population. CONCLUSIONS Cluster analysis using baseline data in ECLIPSE identified five COPD subgroups that differ in outcomes and inflammatory biomarkers and show different relationships between clinical parameters, suggesting the clusters represent clinically and biologically different subtypes of COPD.
Collapse
|
304
|
Lindenmaier TJ, Kirby M, Paulin G, Mielniczuk L, Cunningham IA, Mura M, Licskai C, Parraga G. Pulmonary Artery Abnormalities in Ex-smokers with and without Airflow Obstruction. COPD 2015; 13:224-34. [DOI: 10.3109/15412555.2015.1074666] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
305
|
Obeidat M, Fishbane N, Nie Y, Chen V, Hollander Z, Tebbutt SJ, Bossé Y, Ng RT, Miller BE, McManus B, Rennard S, Paré PD, Sin DD. The Effect of Statins on Blood Gene Expression in COPD. PLoS One 2015; 10:e0140022. [PMID: 26462087 PMCID: PMC4604084 DOI: 10.1371/journal.pone.0140022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 09/19/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COPD is currently the fourth leading cause of death worldwide. Statins are lipid lowering agents with documented cardiovascular benefits. Observational studies have shown that statins may have a beneficial role in COPD. The impact of statins on blood gene expression from COPD patients is largely unknown. OBJECTIVE Identify blood gene signature associated with statin use in COPD patients, and the pathways underpinning this signature that could explain any potential benefits in COPD. METHODS Whole blood gene expression was measured on 168 statin users and 451 non-users from the ECLIPSE study using the Affymetrix Human Gene 1.1 ST microarray chips. Factor Analysis for Robust Microarray Summarization (FARMS) was used to process the expression data. Differential gene expression analysis was undertaken using the Linear Models for Microarray data (Limma) package adjusting for propensity score and surrogate variables. Similarity of the expression signal with published gene expression profiles was performed in ProfileChaser. RESULTS 25 genes were differentially expressed between statin users and non-users at an FDR of 10%, including LDLR, CXCR2, SC4MOL, FAM108A1, IFI35, FRYL, ABCG1, MYLIP, and DHCR24. The 25 genes were significantly enriched in cholesterol homeostasis and metabolism pathways. The resulting gene signature showed correlation with Huntington's disease, Parkinson's disease and acute myeloid leukemia gene signatures. CONCLUSION The blood gene signature of statins' use in COPD patients was enriched in cholesterol homeostasis pathways. Further studies are needed to delineate the role of these pathways in lung biology.
Collapse
Affiliation(s)
- Ma’en Obeidat
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Nick Fishbane
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Yunlong Nie
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
| | - Virginia Chen
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Zsuzsanna Hollander
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Scott J. Tebbutt
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Department of Molecular Medicine, Laval University, Québec, Canada
| | - Raymond T. Ng
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Bruce E. Miller
- Respiratory Therapy Area Unit, GlaxoSmithKline R&D, King of Prussia, Pennsylvania, United States of America
| | - Bruce McManus
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Prevention of Organ Failure (PROOF) Centre of Excellence, Vancouver, BC, Canada
| | - Stephen Rennard
- Division of Pulmonary and Critical Care Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Peter D. Paré
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Don D. Sin
- The University of British Columbia Centre for Heart Lung Innovation, St Paul’s Hospital, Vancouver, BC, Canada
- Respiratory Division, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
306
|
Exuzides A, Colby C, Briggs AH, Lomas DA, Rutten-van Mölken MPMH, Tabberer M, Chambers M, Muellerova H, Locantore N, Risebrough NA, Ismaila AS, Gonzalez-McQuire S. Statistical Modeling of Disease Progression for Chronic Obstructive Pulmonary Disease Using Data from the ECLIPSE Study. Med Decis Making 2015; 37:453-468. [PMID: 26449490 DOI: 10.1177/0272989x15610781] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND To develop statistical models predicting disease progression and outcomes in chronic obstructive pulmonary disease (COPD), using data from ECLIPSE, a large, observational study of current and former smokers with COPD. METHODS Based on a conceptual model of COPD disease progression and data from 2164 patients, associations were made between baseline characteristics, COPD disease progression attributes (exacerbations, lung function, exercise capacity, and symptoms), health-related quality of life (HRQoL), and survival. Linear and nonlinear functional forms of random intercept models were used to characterize these relationships. Endogeneity was addressed by time-lagging variables in the regression models. RESULTS At the 5% significance level, an exacerbation history in the year before baseline was associated with increased risk of future exacerbations (moderate: +125.8%; severe: +89.2%) and decline in lung function (forced expiratory volume in 1 second [FEV1]) (-94.20 mL per year). Each 1% increase in FEV1 % predicted was associated with decreased risk of exacerbations (moderate: -1.1%; severe: -3.0%) and increased 6-minute walk test distance (6MWD) (+1.5 m). Increases in baseline exercise capacity (6MWD, per meter) were associated with slightly increased risk of moderate exacerbations (+0.04%) and increased FEV1 (+0.62 mL). Symptoms (dyspnea, cough, and/or sputum) were associated with an increased risk of moderate exacerbations (+13.4% to +31.1%), and baseline dyspnea (modified Medical Research Council score ≥2 v. <2) was associated with lower FEV1 (-112.3 mL). CONCLUSIONS A series of linked statistical regression equations have been developed to express associations between indicators of COPD disease severity and HRQoL and survival. These can be used to represent disease progression, for example, in new economic models of COPD.
Collapse
Affiliation(s)
| | | | - Andrew H Briggs
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK (AHB).,ICON Health Economics, Morristown, NJ, USA (AHB)
| | - David A Lomas
- Wolfson Institute for Biomedical Research, University College London, London, UK (DAL)
| | | | - Maggie Tabberer
- Value Evidence and Outcomes, GSK R&D, Uxbridge, UK (MT, SG-M)
| | - Mike Chambers
- Global Market Access and Healthcare Solutions, GSK, Brentford, UK (MC)
| | | | | | | | - Afisi S Ismaila
- Value Evidence and Outcomes, GSK R&D, Research Triangle Park, NC, USA (ASI).,Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, ON, Canada (ASI)
| | | |
Collapse
|
307
|
Ambrosino N, Casaburi R, Chetta A, Clini E, Donner CF, Dreher M, Goldstein R, Jubran A, Nici L, Owen CA, Rochester C, Tobin MJ, Vagheggini G, Vitacca M, ZuWallack R. 8th international conference on management and rehabilitation of chronic respiratory failure: the long summaries – part 1. Multidiscip Respir Med 2015. [PMCID: PMC4595244 DOI: 10.1186/s40248-015-0026-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This paper summarizes the Part 1 of the proceedings of the 8th International Conference on Management and Rehabilitation of Chronic Respiratory Failure, held in Pescara, Italy, on 7 and 8 May, 2015. It summarizes the contributions from numerous experts in the field of chronic respiratory disease and chronic respiratory failure. The outline follows the temporal sequence of presentations. This paper (Part 1) includes sections regarding: Advances in Asthma and COPD Therapy (Novel Therapeutic Targets for Asthma: Proteinases, Blood Biomarker Changes in COPD Patients); The problem of Hospital Re-Admission following Discharge after the COPD Exacerbation (Characteristics of the Hospitalized COPD Patient, Reducing Hospital Readmissions Following COPD Exacerbation).
Collapse
|
308
|
Titz B, Sewer A, Schneider T, Elamin A, Martin F, Dijon S, Luettich K, Guedj E, Vuillaume G, Ivanov NV, Peck MJ, Chaudhary NI, Hoeng J, Peitsch MC. Alterations in the sputum proteome and transcriptome in smokers and early-stage COPD subjects. J Proteomics 2015; 128:306-20. [DOI: 10.1016/j.jprot.2015.08.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/15/2015] [Indexed: 12/15/2022]
|
309
|
Katajisto M, Koskela J, Lindqvist A, Kilpeläinen M, Laitinen T. Physical activity in COPD patients decreases short-acting bronchodilator use and the number of exacerbations. Respir Med 2015; 109:1320-5. [DOI: 10.1016/j.rmed.2015.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 07/07/2015] [Accepted: 08/02/2015] [Indexed: 10/23/2022]
|
310
|
Rutten EPA, Spruit MA, McDonald MLN, Rennard S, Agusti A, Celli B, Miller BE, Crim C, Calverley PMA, Hanson C, MacNee W, Franssen FME, Vanfleteren L, Wouters EFM. Continuous fat-free mass decline in COPD: fact or fiction? Eur Respir J 2015; 46:1496-8. [PMID: 26381518 DOI: 10.1183/13993003.00692-2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/09/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Erica P A Rutten
- Dept of Research and Education, CIRO, Horn, The Netherlands Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martijn A Spruit
- Dept of Research and Education, CIRO, Horn, The Netherlands REVAL - Rehabilitation Research Center, BIOMED - Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Merry-Lynn N McDonald
- Channing Division of Network Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Stephen Rennard
- Division of Internal Medicine, University of Nebraska, Nebraska Medical Center, Omaha, NE, USA
| | - Alvar Agusti
- Thorax Institute, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain CIBERES, Madrid, Spain
| | - Bartolome Celli
- Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Corrine Hanson
- Medical Nutrition Education, University of Nebraska, Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | - Emiel F M Wouters
- Dept of Research and Education, CIRO, Horn, The Netherlands Maastricht University Medical Center, Maastricht, The Netherlands
| | | |
Collapse
|
311
|
Rooney C, Sethi T. Biomarkers for precision medicine in airways disease. Ann N Y Acad Sci 2015; 1346:18-32. [PMID: 26099690 DOI: 10.1111/nyas.12809] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 05/12/2015] [Accepted: 05/13/2015] [Indexed: 12/22/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex clinical entity. In contrast to previously limited diagnostic definitions, it is now apparent that COPD is a clinically and biologically heterogeneous disease process, overlapping with other airways diseases like chronic asthma. As such, symptomatic response to current standard treatment practices is variable. New clinical guidelines have been altered to reflect this, with the inclusion of symptoms and risk factors in diagnostic and management algorithms. However, as our understanding of COPD pathophysiology deepens, many novel physiological, cellular, proteomic, and genetic markers have been identified. Several have been observed to be independently predictive of distinct clinical disease patterns, which at present are not illustrated by conventional measurements of lung impairment. The potential use of these predictive biomarkers to stratify this diverse patient population could transform the care we offer. We should aim for precision medicine to optimize diagnosis and treatment choices and to monitor and improve clinical outcomes in this disease.
Collapse
Affiliation(s)
| | - Tariq Sethi
- Asthma, Allergy and Lung Biology, King's College London, London, United Kingdom
| |
Collapse
|
312
|
Cho MH, Castaldi PJ, Hersh CP, Hobbs BD, Barr RG, Tal-Singer R, Bakke P, Gulsvik A, San José Estépar R, Van Beek EJR, Coxson HO, Lynch DA, Washko GR, Laird NM, Crapo JD, Beaty TH, Silverman EK. A Genome-Wide Association Study of Emphysema and Airway Quantitative Imaging Phenotypes. Am J Respir Crit Care Med 2015; 192:559-69. [PMID: 26030696 PMCID: PMC4595690 DOI: 10.1164/rccm.201501-0148oc] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 05/28/2015] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is defined by the presence of airflow limitation on spirometry, yet subjects with COPD can have marked differences in computed tomography imaging. These differences may be driven by genetic factors. We hypothesized that a genome-wide association study (GWAS) of quantitative imaging would identify loci not previously identified in analyses of COPD or spirometry. In addition, we sought to determine whether previously described genome-wide significant COPD and spirometric loci were associated with emphysema or airway phenotypes. OBJECTIVES To identify genetic determinants of quantitative imaging phenotypes. METHODS We performed a GWAS on two quantitative emphysema and two quantitative airway imaging phenotypes in the COPDGene (non-Hispanic white and African American), ECLIPSE (Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints), NETT (National Emphysema Treatment Trial), and GenKOLS (Genetics of COPD, Norway) studies and on percentage gas trapping in COPDGene. We also examined specific loci reported as genome-wide significant for spirometric phenotypes related to airflow limitation or COPD. MEASUREMENTS AND MAIN RESULTS The total sample size across all cohorts was 12,031, of whom 9,338 were from COPDGene. We identified five loci associated with emphysema-related phenotypes, one with airway-related phenotypes, and two with gas trapping. These loci included previously reported associations, including the HHIP, 15q25, and AGER loci, as well as novel associations near SERPINA10 and DLC1. All previously reported COPD and a significant number of spirometric GWAS loci were at least nominally (P < 0.05) associated with either emphysema or airway phenotypes. CONCLUSIONS Genome-wide analysis may identify novel risk factors for quantitative imaging characteristics in COPD and also identify imaging features associated with previously identified lung function loci.
Collapse
Affiliation(s)
- Michael H. Cho
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Craig P. Hersh
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Brian D. Hobbs
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - R. Graham Barr
- Department of Medicine, College of Physicians and Surgeons, and
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| | - Ruth Tal-Singer
- GlaxoSmithKline Research and Development, King of Prussia, Pennsylvania
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Amund Gulsvik
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Raúl San José Estépar
- Laboratory of Mathematics in Imaging, Department of Radiology, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Edwin J. R. Van Beek
- Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- Department of Radiology and
- Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa
| | - Harvey O. Coxson
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| | - David A. Lynch
- Department of Radiology, National Jewish Health, Denver, Colorado
| | - George R. Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Nan M. Laird
- Harvard School of Public Health, Boston, Massachusetts; and
| | - James D. Crapo
- Department of Radiology, National Jewish Health, Denver, Colorado
| | - Terri H. Beaty
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Edwin K. Silverman
- Channing Division of Network Medicine and
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| |
Collapse
|
313
|
Sheikh K, Coxson HO, Parraga G. This
is what
COPD
looks like. Respirology 2015; 21:224-36. [DOI: 10.1111/resp.12611] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 06/22/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Khadija Sheikh
- Robarts Research Institute London Canada
- Department of Medical BiophysicsThe University of Western Ontario London Canada
| | - Harvey O Coxson
- UBC Centre for Heart Lung InnovationSt. Paul's Hospital Vancouver Canada
- Department of RadiologyUniversity of British Columbia Vancouver Canada
| | - Grace Parraga
- Robarts Research Institute London Canada
- Department of Medical BiophysicsThe University of Western Ontario London Canada
| |
Collapse
|
314
|
Goldman AW, Burmeister Y, Cesnulevicius K, Herbert M, Kane M, Lescheid D, McCaffrey T, Schultz M, Seilheimer B, Smit A, St Laurent G, Berman B. Bioregulatory systems medicine: an innovative approach to integrating the science of molecular networks, inflammation, and systems biology with the patient's autoregulatory capacity? Front Physiol 2015; 6:225. [PMID: 26347656 PMCID: PMC4541032 DOI: 10.3389/fphys.2015.00225] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/27/2015] [Indexed: 12/25/2022] Open
Abstract
Bioregulatory systems medicine (BrSM) is a paradigm that aims to advance current medical practices. The basic scientific and clinical tenets of this approach embrace an interconnected picture of human health, supported largely by recent advances in systems biology and genomics, and focus on the implications of multi-scale interconnectivity for improving therapeutic approaches to disease. This article introduces the formal incorporation of these scientific and clinical elements into a cohesive theoretical model of the BrSM approach. The authors review this integrated body of knowledge and discuss how the emergent conceptual model offers the medical field a new avenue for extending the armamentarium of current treatment and healthcare, with the ultimate goal of improving population health.
Collapse
Affiliation(s)
- Alyssa W Goldman
- Concept Systems, Inc. Ithaca, NY, USA ; Department of Sociology, Cornell University Ithaca, NY, USA
| | | | | | - Martha Herbert
- Transcend Research Laboratory, Massachusetts General Hospital Boston, MA, USA
| | - Mary Kane
- Concept Systems, Inc. Ithaca, NY, USA
| | - David Lescheid
- International Academy of Bioregulatory Medicine Baden-Baden, Germany
| | - Timothy McCaffrey
- Division of Genomic Medicine, George Washington University Medical Center Washington, DC, USA
| | - Myron Schultz
- Biologische Heilmittel Heel GmbH Baden-Baden, Germany
| | | | - Alta Smit
- Biologische Heilmittel Heel GmbH Baden-Baden, Germany
| | | | - Brian Berman
- Center for Integrative Medicine, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
315
|
Malinovschi A, Ludviksdottir D, Tufvesson E, Rolla G, Bjermer L, Alving K, Diamant Z. Application of nitric oxide measurements in clinical conditions beyond asthma. Eur Clin Respir J 2015; 2:28517. [PMID: 26672962 PMCID: PMC4653314 DOI: 10.3402/ecrj.v2.28517] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/05/2015] [Indexed: 02/01/2023] Open
Abstract
Fractional exhaled nitric oxide (FeNO) is a convenient, non-invasive method for the assessment of active, mainly Th2-driven, airway inflammation, which is sensitive to treatment with standard anti-inflammatory therapy. Consequently, FeNO serves as a valued tool to aid diagnosis and monitoring in several asthma phenotypes. More recently, FeNO has been evaluated in several other respiratory, infectious, and/or immunological conditions. In this short review, we provide an overview of several clinical studies and discuss the status of potential applications of NO measurements in clinical conditions beyond asthma.
Collapse
Affiliation(s)
- Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden;
| | - Dora Ludviksdottir
- Department of Respiratory Medicine and Sleep, Landspitali University Hospital, Reykjavik, Iceland
| | - Ellen Tufvesson
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Lund University, Lund, Sweden
| | - Giovanni Rolla
- Department of Medical Sciences, Allergology and Clinical Immunology, University of Torino, Torino, Italy
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Lund University, Lund, Sweden
| | - Kjell Alving
- Department of Women's and Children's Health, Uppsala University, Uppsala, Sweden
| | - Zuzana Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Lund University, Lund, Sweden.,Department of Clinical Pharmacy & Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands.,Department of General Practice, University Medical Centre Groningen, Groningen, The Netherlands.,QPS Netherlands, Groningen, The Netherlands
| |
Collapse
|
316
|
Di Marco F, Santus P, Sotgiu G, Blasi F, Centanni S. Does Improving Exercise Capacity and Daily Activity Represent the Holistic Perspective of a New COPD Approach? COPD 2015; 12:575-81. [PMID: 26457460 DOI: 10.3109/15412555.2015.1008694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In COPD patients a reduced daily activity has been well documented, resulting from both respiratory and non-respiratory manifestations of the disease. An evaluation by multisensory armband has confirmed that daily physical activity is mainly associated with dynamic hyperinflation, regardless of COPD severity. This aspect is crucial, since exercise capacity is closely correlated to life expectancy. Notwithstanding the causal key role of lung impairment in the patient's symptoms, some authors have suggested that other factors, such as systemic inflammation and co-morbidities, have an important role, particularly as mortality risk factors. Many studies suggest the efficacy of bronchodilators and rehabilitation in improving exercise capacity, and, speaking in terms of daily life, in increasing the number of days in which patients are able to perform their usual activities. On this evidence, the first aim in the management of COPD should be to improve exercise capacity and daily activity since these outcomes have direct effects on patients' quality of life, co-morbidities (heart and metabolic diseases), and prognosis. Thus, improving physical activity represents a modern approach aimed at dealing with both pulmonary and systemic manifestations of the disease. It is however worth of notice to remember that in patients affected by COPD the relationship between the improvement of "potential" exercise capacity and daily physical activity has been found to be only moderate to weak. Obtaining a significant behavior modification with regard to daily physical activity, together with the optimization of therapy thus represents currently the true challenge.
Collapse
Affiliation(s)
- Fabiano Di Marco
- a Respiratory Unit, Ospedale San Paolo, Dept of Scienze della Salute , Università degli Studi di Milano , Milan , Italy
| | - Pierachille Santus
- b Respiratory Unit Fondazione Salvatore Maugeri-Istituto Scientifico di Milano - IRCCS , Università degli Studi di Milano , Milan , Italy
| | - Giovanni Sotgiu
- c Clinical Epidemiology and Medical Statistics Unit, Dept of Biomedical Sciences , University of Sassari - Research, Medical Education and Professional Development Unit , AOU Sassari , Italy
| | - Francesco Blasi
- d Respiratory Unit, IRCCS Fondazione Cà Granda Milano, Department of Pathophysiology and Transplantation , University of Milan , Milan , Italy
| | - Stefano Centanni
- a Respiratory Unit, Ospedale San Paolo, Dept of Scienze della Salute , Università degli Studi di Milano , Milan , Italy
| |
Collapse
|
317
|
Setoguchi Y, Izumi S, Nakamura H, Hanada S, Marumo K, Kurosaki A, Akata S. Survey to determine the efficacy and safety of guideline-based pharmacological therapy for chronic obstructive pulmonary disease patients not previously receiving maintenance treatment. Expert Opin Pharmacother 2015; 16:2271-81. [PMID: 26290277 DOI: 10.1517/14656566.2015.1074678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To investigate the potential beneficial effects of guideline-based pharmacological therapy on pulmonary function and quality of life (QOL) in Japanese chronic obstructive pulmonary disease (COPD) patients without prior treatment. RESEARCH DESIGN AND METHODS Multicenter survey, open-label study of 49 Japanese COPD patients aged ≥ 40 years; outpatients with >10 pack years of smoking history; ratio of forced expiratory volume in 1 s (FEV1)/forced vital capacity (FVC) < 70%; predicted FEV1 < 80%; treated with bronchodilators and/or inhaled corticosteroids as maintenance therapy until week 48. MAIN OUTCOME MEASURES The primary endpoint was change in pulmonary function (trough FEV1, trough FVC); secondary endpoints were QOL and physical activity at 48 weeks after initiation of therapy. RESULTS Airway reversibility was confirmed in untreated patients. Significant changes over time were not observed for FEV1 and FVC, indicating lung function at initiation of treatment was maintained during the observation period. COPD assessment test scores showed statistical and clinical improvements. Cough, sputum, breathlessness, and shortness of breath were significantly improved. CONCLUSIONS Lung function and QOL of untreated Japanese COPD patients improved and improvements were maintained by performing a therapeutic intervention that conformed to published guidelines.
Collapse
Affiliation(s)
- Yasuhiro Setoguchi
- a 1 Tokyo Medical University, Department of Respiratory Medicine , 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan +81 333 426 111 ; +81 333 425 070 ;
| | - Shinyu Izumi
- b 2 National Center for Global Health and Medicine, Department of Respiratory Medicine , Tokyo, Japan
| | - Hidenori Nakamura
- c 3 Seirei Hamamatsu General Hospital, Department of Respiratory Medicine , Shizuoka, Japan
| | - Shigeo Hanada
- d 4 Respiratory Center, Toranomon Hospital, Department of Respiratory Medicine , Tokyo, Japan
| | - Kazuyoshi Marumo
- e 5 Tokyo Metropolitan Police Hospital, Department of Internal Medicine , Tokyo, Japan
| | - Atsuko Kurosaki
- f 6 Fukujuji Hospital, Department of Diagnostic Radiology , Tokyo, Japan
| | - Shouichi Akata
- g 7 Tokyo Medical University, Department of Radiology , Tokyo, Japan
| |
Collapse
|
318
|
Come CE, Washko GR. CT Scanning in COPD - Is it Time to Move On? CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2015; 2:201-203. [PMID: 28848843 DOI: 10.15326/jcopdf.2.3.2015.0150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Carolyn E Come
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
319
|
Kirby M, Pike D, Sin DD, Coxson HO, McCormack DG, Parraga G. COPD: Do Imaging Measurements of Emphysema and Airway Disease Explain Symptoms and Exercise Capacity? Radiology 2015; 277:872-80. [PMID: 26151081 DOI: 10.1148/radiol.2015150037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To determine the role of imaging measurements of emphysema and airway disease in determining chronic obstructive pulmonary disease (COPD) symptoms and exercise limitation in patients with COPD, particularly in patients with mild-to-moderate disease. MATERIALS AND METHODS Participants (n = 116) with Global Initiative for Chronic Obstructive Lung Disease (GOLD) grade U (unclassified) or grade I-IV COPD provided informed consent to an ethics board-approved HIPAA-compliant protocol and underwent spirometry and plethysmography, completed the St George's Respiratory Questionnaire (SGRQ), completed a 6-minute walk test for the 6-minute walk distance (6MWD), and underwent hyperpolarized helium 3 ((3)He) magnetic resonance (MR) imaging and computed tomography (CT). Emphysema was estimated by using the MR imaging apparent diffusion coefficient (ADC) and the relative area of the CT attenuation histogram with attenuation of -950 HU or less (RA950). Airway disease was measured by using the CT airway wall thickness of airways with an internal perimeter of 10 mm and total airway count. Ventilation defect percentage at (3)He MR imaging was used to measure ventilation. Multivariable regression models for the 6MWD and SGRQ symptom subscore were used to evaluate the relationships between physiologic and imaging measurements. RESULTS Multivariate modeling for the 6MWD in 80 patients with GOLD grade U-II COPD showed that ADC (β = 0.34, P = .04), diffusing capacity of the lung for carbon monoxide (β = 0.60, P = .0008), and residual volume/total lung capacity (β = -0.26, P = .02) were significant variables, while forced expiratory volume in 1 second (FEV1) and airway disease measurements were not. In 36 patients with GOLD grade III or IV disease, FEV1 (β = 0.48, P = .01) was the only significant contributor in a multivariate model for 6MWD. MR imaging emphysema measurements also made the greatest relative contribution to symptoms in patients with milder (GOLD grade U-II) COPD (ADC: β = 0.60, P = .005; RA950: β = -0.52, P = .02; FEV1: β = -0.45, P = .0002) and in grade III or IV disease (ADC: β = 0.95, P = .01; RA950: β = -0.62, P = .07; airway count: β = -0.49, P = .01). CONCLUSION In patients with mild-to-moderate COPD, MR imaging emphysema measurements played a dominant role in the expression of exercise limitation, while both CT and MR imaging measurements of emphysema explained symptoms.
Collapse
Affiliation(s)
- Miranda Kirby
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| | - Damien Pike
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| | - Don D Sin
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| | - Harvey O Coxson
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| | - David G McCormack
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| | - Grace Parraga
- From the James Hogg Research Centre, the University of British Columbia and the Institute of Heart and Lung Health, St Paul's Hospital, Vancouver, BC, Canada (M.K., D.D.S., H.O.C.); Imaging Research Laboratories, Robarts Research Institute (D.P., G.P.), and Department of Medical Biophysics (D.P., G.P.) and Division of Respirology, Department of Medicine (D.G.M.), the University of Western Ontario, 1151 Richmond St N, London, ON, Canada N6A 5B7
| |
Collapse
|
320
|
Bihlet AR, Karsdal MA, Bay-Jensen AC, Read S, Kristensen JH, Sand JMB, Leeming DJ, Andersen JR, Lange P, Vestbo J. Clinical Drug Development Using Dynamic Biomarkers to Enable Personalized Health Care in COPD. Chest 2015; 148:16-23. [DOI: 10.1378/chest.15-0296] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
321
|
Prognostic value of variables derived from the six-minute walk test in patients with COPD: Results from the ECLIPSE study. Respir Med 2015; 109:1138-46. [PMID: 26143282 DOI: 10.1016/j.rmed.2015.06.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 05/28/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
In addition to the six-min walk distance (6 MWD), other six-min walk test (6 MWT) derived variables, such as mean walk-speed (6MWSpeed), 6-min walk-work (6 MWW), distance-saturation product (DSP), exercise-induced oxygen desaturation (EID), and unintended stops may be useful for the prediction of mortality and hospitalization in patients with chronic obstructive pulmonary disease (COPD). We studied the association between 6 MWT-derived variables and mortality as well as hospitalization in COPD patients and compared it with the BODE index. A three-year prospective study (ECLIPSE) to evaluate the prognostic value of 6 MWT-derived variables in 2010 COPD patients. Cox's proportional-hazard regressions were performed to estimate 3-year mortality and hospitalization. During the follow-up, 193 subjects died and 622 were hospitalized. An adjusted Cox's regression model of hazard ratio [HR] for impaired 6 MWT-derived variables was significant referring to: mortality (6 MWD ≤334 m [2.30], 6MWSpeed ≤0.9 m/sec [2.15], 6 MWW ≤20000 m kg [2.17], DSP ≤290 m% [2.70], EID ≤88% [1.75], unintended stops [1.99]; and hospitalization (6 MWW ≤27000 m kg [1.23], EID ≤88% [1.25], BODE index ≥3 points [1.40]; all p ≤ 0.05). The 6 MWT-derived variables have an additional predictive value of mortality in patients with COPD. The 6 MWW, EID and the BODE index refine the prognosis of hospitalization.
Collapse
|
322
|
Benson VS, Müllerová H, Vestbo J, Wedzicha JA, Patel A, Hurst JR. Associations between gastro-oesophageal reflux, its management and exacerbations of chronic obstructive pulmonary disease. Respir Med 2015; 109:1147-54. [PMID: 26166017 DOI: 10.1016/j.rmed.2015.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/18/2015] [Accepted: 06/17/2015] [Indexed: 01/06/2023]
Abstract
AIM To determine factors, overall and by sex, associated with self-reported gastro-oesophageal reflux disease (GORD) in chronic obstructive pulmonary disease (COPD) patients, and to evaluate relationships between GORD, its modification by acid suppression medications (Proton Pump Inhibitors [PPI]/histamine-2 receptor antagonists [H2RA]) and exacerbations of COPD and mortality. METHODS Logistic regression was used to determine factors associated with GORD; Cox proportional hazards models were used to calculate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for GORD and risk of exacerbation and death. RESULTS Among 2135 COPD patients from the ECLIPSE cohort, 547 patients self-reported GORD, with female preponderance; 237 were taking PPI/H2RA. Risk factors for GORD did not differ by sex. When compared to patients who did not report GORD or use of PPI/H2RA, patients with GORD and taking PPI/H2RA had a significantly increased risk of exacerbation (HR = 1.58, 95%CI = 1.35-1.86); risk was also increased for patients reporting GORD only or PPI/H2RA use only (HR = 1.21 [1.04-1.40] and 1.33 [1.08-1.65], respectively). Similar findings were observed for risk of hospitalised exacerbation. GORD was not associated with mortality. CONCLUSION GORD in COPD patients is highly prevalent, and risk factors did not differ by sex. Use of PPI/H2RA and self-reported GORD were associated with increased risk of moderate-to-severe and hospitalised exacerbations.
Collapse
Affiliation(s)
| | - Hana Müllerová
- Respiratory Epidemiology, GSK R&D, Uxbridge, United Kingdom
| | - Jørgen Vestbo
- Centre for Respiratory and Medicine and Allergy, Institute of Inflammation and Repair, Manchester Academic Health Science Centre, The University of Manchester and University Hospital South Manchester NHS Foundation Trust, Manchester, United Kingdom
| | - Jadwiga A Wedzicha
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Anant Patel
- UCL Respiratory Medicine, University College London, Royal Free Campus, Rowland Hill Street, London, United Kingdom
| | - John R Hurst
- UCL Respiratory Medicine, University College London, Royal Free Campus, Rowland Hill Street, London, United Kingdom.
| | | |
Collapse
|
323
|
Müllerova H, Maselli DJ, Locantore N, Vestbo J, Hurst JR, Wedzicha JA, Bakke P, Agusti A, Anzueto A. Hospitalized exacerbations of COPD: risk factors and outcomes in the ECLIPSE cohort. Chest 2015; 147:999-1007. [PMID: 25356881 DOI: 10.1378/chest.14-0655] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Exacerbations of COPD requiring hospital admission have important clinical and societal implications. We sought to investigate the incidence, recurrence, risk factors, and mortality of patients with COPD exacerbations requiring hospital admission compared with those without hospital admission during 3-year follow-up. Patients with COPD (N = 2,138) were identified from the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) observational cohort. METHODS An analysis of time to first event of hospital admission was performed using Kaplan-Meier curves and Cox proportional hazard regression adjusting for possible confounders. RESULTS Of the 2,138 patients, 670 (31%) reported a total of 1,452 COPD exacerbations requiring hospital admission during the study period; 313 patients (15%) reported multiple events. A prior history of exacerbation of COPD requiring hospital admission was the factor associated with the highest risk of a new hospitalization for exacerbation (hazard ratio, 2.71; 95% CI, 2.24-3.29; P < .001). Other risk factors included more severe airflow limitation, poorer health status, older age, radiologic evidence of emphysema, and higher WBC count. Having been hospitalized for exacerbation significantly increased the risk of mortality (P < .001). CONCLUSIONS Exacerbations of COPD requiring hospital admission occur across all stages of airflow limitation and are a significant prognostic factor of reduced survival across all COPD stages. Patients with COPD at a high risk for hospitalization can be identified by their past history for similar events, and other factors, including the severity of airflow limitation, poor health status, age, presence of emphysema, and leukocytosis. TRIAL REGISTRY ClinicalTrials.gov; No.: NCT00292552; URL: www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Hana Müllerova
- Respiratory Epidemiology, GlaxoSmithKline R&D, Uxbridge, England
| | - Diego J Maselli
- Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, TX; Division of Pulmonary Diseases/Critical Care Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Nicholas Locantore
- Respiratory Medicines Development Center, GlaxoSmithKline, Research Triangle Park, NC
| | - Jørgen Vestbo
- Gentofte, Hellerup, Denmark; Respiratory Research Group, Manchester Academic Health Science Centre, University of Manchester, Manchester, England
| | - John R Hurst
- Centre of Inflammation and Tissue Repair, University College London, London, England
| | - Jadwiga A Wedzicha
- Centre for Respiratory Medicine, University College London, London, England
| | - Per Bakke
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Alvar Agusti
- Thorax Institute, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain; FISIB, CIBER Enfermedades Respiratorias, Mallorca, Spain
| | - Antonio Anzueto
- Audie L. Murphy Hospital, South Texas Veterans Health Care System, San Antonio, TX; Division of Pulmonary Diseases/Critical Care Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX.
| |
Collapse
|
324
|
Lopez-Campos JL, Fernandez-Villar A, Calero-Acuña C, Represas-Represas C, Lopez-Ramírez C, Fernández VL, Soler-Cataluña JJ, Casamor R. Evaluation of the COPD Assessment Test and GOLD patient types: a cross-sectional analysis. Int J Chron Obstruct Pulmon Dis 2015; 10:975-84. [PMID: 26064045 PMCID: PMC4455854 DOI: 10.2147/copd.s82781] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The COPD Assessment Test (CAT) has been recently developed to quantify COPD impact in routine practice. However, no relationship with other measures in the Global Initiative for Obstructive Lung Disease (GOLD) strategy has been evaluated. The present study aimed to evaluate the relationship of the CAT with other GOLD multidimensional axes, patient types, and the number of comorbidities. Methods This was a cross-sectional analysis of the Clinical presentation, diagnosis, and course of chronic obstructive pulmonary disease (On-Sint) study. The CAT score was administered to all participants at the inclusion visit. A GOLD 2011 strategy consisting of modified Medical Research Council scale (MRC) scores was devised to study the relationship between the CAT, and GOLD 2011 axes and patient types. The relationship with comorbidities was assessed using the Charlson comorbidity index, grouped as zero, one to two, and three or more. Results The CAT questionnaire was completed by 1,212 patients with COPD. The CAT maintained a relationship with all the three axes, with a ceiling effect for dyspnea and no distinction between mild and moderate functional impairment. The CAT score increased across GOLD 2011 patient types A–D, with similar scores for types B and C. Within each GOLD 2011 patient type, there was a considerably wide distribution of CAT values. Conclusion Our study indicates a correlation between CAT and the GOLD 2011 classification axes as well as the number of comorbidities. The CAT score can help clinicians, as a complementary tool to evaluate patients with COPD within the different GOLD patient types.
Collapse
Affiliation(s)
- Jose Luis Lopez-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/Universidad de Sevilla, Sevilla, Spain ; CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Fernandez-Villar
- Servicio de Neumología, Instituto de Investigación Biomédica de Vigo (IBIV), Complexo Hospitalario de Vigo, Vigo, Spain
| | - Carmen Calero-Acuña
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/Universidad de Sevilla, Sevilla, Spain
| | - Cristina Represas-Represas
- Servicio de Neumología, Instituto de Investigación Biomédica de Vigo (IBIV), Complexo Hospitalario de Vigo, Vigo, Spain
| | - Cecilia Lopez-Ramírez
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/Universidad de Sevilla, Sevilla, Spain
| | - Virginia Leiro Fernández
- Servicio de Neumología, Instituto de Investigación Biomédica de Vigo (IBIV), Complexo Hospitalario de Vigo, Vigo, Spain
| | - Juan Jose Soler-Cataluña
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain ; Servicio de Neumología Hospital Arnau de Vilanova, Valencia, Spain
| | - Ricard Casamor
- Departamento Médico de Novartis Farmacéutica, Barcelona, Spain
| | | |
Collapse
|
325
|
Thomsen LH, Shaker SB, Dirksen A, Pedersen JH, Tal-Singer R, Bakke P, Vestbo J. Correlation Between Emphysema and Lung Function in Healthy Smokers and Smokers With COPD. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2015; 2:204-213. [PMID: 28848844 DOI: 10.15326/jcopdf.2.3.2014.0154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background: Emphysema is an important component of COPD; however, in previous studies of the correlation between airflow limitation (AFL) and computed tomography (CT) lung density as a surrogate for emphysema has varied. We hypothesised a good correlation between lung function (forced expiratory volume in first second [FEV1]) and emphysema (15th percentile density [PD15]) and that this correlation also exists between loss of lung tissue and decline in lung function even within the time frame of longitudinal studies of relatively short duration. Methods: We combined 2 large longitudinal studies (the Danish Lung Cancer Screening Trial [DLCST] and the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints [ECLIPSE]) of smokers or former smokers, with a wide range of AFL and CT lung density, and analysed data from 2148 participants who did not change smoking habits and who had at least 2 CT scans and 2 FEV1 measurements at least 3 years apart. Results: Baseline correlation between FEV1 and PD15 was high (r=0.716, 95% confidence interval [CI]: 0.694-0.736, p<0.001) indicating that at least half of the variation in FEV1 can be explained by variation in CT lung density. Correlation between the decline in FEV1 and progression of PD15 was considerably weaker (r= 0.081, 95% CI: 0.038-0.122, p<0.001). Conclusions: Correlation is very high between lung density and lung function in a broad spectrum of smokers and ex-smokers. In contrast, the temporal associations (slopes) are weakly correlated, probably due to uncertainty in the estimation of slopes within a time frame of 3-4 years.
Collapse
Affiliation(s)
- Laura H Thomsen
- Department of Respiratory Medicine, Gentofte Hospital, University of Copenhagen, Denmark
| | - Saher B Shaker
- Department of Respiratory Medicine, Gentofte Hospital, University of Copenhagen, Denmark
| | - Asger Dirksen
- Department of Respiratory Medicine, Gentofte Hospital, University of Copenhagen, Denmark
| | - Jesper H Pedersen
- Department of Cardiothoracic Surgery, University of Copenhagen, Denmark
| | | | - Per Bakke
- Department of Clinical Science, University of Bergen, and Department of Thoracic Medicine, Haukeland University Hospital, Bergen, Norway
| | - Jørgen Vestbo
- Department of Respiratory Medicine, Gentofte Hospital, University of Copenhagen, Denmark.,Respiratory and Allergy Research Group, Manchester Academic Health Science Centre, University Hospital South Manchester; NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
326
|
Zhou Y, Bruijnzeel PLB, McCrae C, Zheng J, Nihlen U, Zhou R, Van Geest M, Nilsson A, Hadzovic S, Huhn M, Taib Z, Gu Y, Xie J, Ran P, Chen R, Zhong N. Study on risk factors and phenotypes of acute exacerbations of chronic obstructive pulmonary disease in Guangzhou, China-design and baseline characteristics. J Thorac Dis 2015; 7:720-33. [PMID: 25973239 DOI: 10.3978/j.issn.2072-1439.2015.04.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/13/2015] [Indexed: 11/14/2022]
Abstract
BACKGROUND To describe a study design that focuses on risk factors and patterns of chronic obstructive pulmonary disease (COPD) exacerbations. METHODS A 2-year, single centre, observational study was conducted in Guangzhou in China. The study enrolled 318 subjects with COPD aged 40-79 years, stratified into different but equally sized groups according to global initiative for chronic obstructive lung disease (GOLD) stage (including Stage 0) and 86 lung healthy controls. An assessment each year was scheduled including questionnaires, lung function testing, Chest X-ray and blood collection. A sub-group, called sub-group X, consisting of 203 subjects with COPD and 51 lung healthy controls, was selected to answer a symptom questionnaire daily (EXACT-PRO) via a BlackBerry Personal Digital Assistant (PDA) device. Upon an alert that indicated a change in daily symptom pattern, the patients were contacted by the clinic to decide whether they had experienced an exacerbation and should have an extra visit within 24-48 hours. At an extra visit, nasal and throat swabs, induced sputum and blood were collected. Air pollution, temperature and humidity were also monitored daily. A subset of sub-group X, called sub-group M that consisted of 52 COPD patients and 15 healthy controls was dedicated to measure muscle strength and a dexa scan. RESULTS More than 78% of the enrolled patients completed the study successfully. There appeared a difference between the patient groups and the controls in gender, age, body mass index (BMI), forced expiratory volume in 1 second (FEV1), FEV1/FVC and smoking at baseline. In sub-group X 90 out of 203 (44.4%) selected COPD patients developed one or more exacerbations in the 2-year observation period. They were more severe COPD patients according to GOLD stage at study start. On average most exacerbations occurred in the month March and the least number of exacerbations occurred in October. CONCLUSIONS This study with the obtained patient dataset will allow a better insight in many aspects of exacerbations in COPD (e.g., the identification, the risk factors, phenotypes and the biomarkers).
Collapse
Affiliation(s)
- Yumin Zhou
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Piet L B Bruijnzeel
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Christopher McCrae
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Jinping Zheng
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Ulf Nihlen
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Rong Zhou
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Marleen Van Geest
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Anna Nilsson
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Sinela Hadzovic
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Monika Huhn
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Ziad Taib
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Yi Gu
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Jiaxing Xie
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Pixin Ran
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Rongchang Chen
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| | - Nanshan Zhong
- 1 The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Diseases, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510182, China ; 2 Early Clinical Development, 3 Translational Science, Respiratory, Inflammation and Autoimmunity iMed, AstraZeneca, Mölndal, Sweden ; 4 Translational Science, Asia & Emerging Markets iMed, AstraZeneca, Shanghai 201203, China ; 5 GMED RIA, 6 Statistical Department, 7 Programming Department, AstraZeneca, Mölndal, Sweden
| |
Collapse
|
327
|
Lynch DA, Austin JHM, Hogg JC, Grenier PA, Kauczor HU, Bankier AA, Barr RG, Colby TV, Galvin JR, Gevenois PA, Coxson HO, Hoffman EA, Newell JD, Pistolesi M, Silverman EK, Crapo JD. CT-Definable Subtypes of Chronic Obstructive Pulmonary Disease: A Statement of the Fleischner Society. Radiology 2015; 277:192-205. [PMID: 25961632 DOI: 10.1148/radiol.2015141579] [Citation(s) in RCA: 401] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The purpose of this statement is to describe and define the phenotypic abnormalities that can be identified on visual and quantitative evaluation of computed tomographic (CT) images in subjects with chronic obstructive pulmonary disease (COPD), with the goal of contributing to a personalized approach to the treatment of patients with COPD. Quantitative CT is useful for identifying and sequentially evaluating the extent of emphysematous lung destruction, changes in airway walls, and expiratory air trapping. However, visual assessment of CT scans remains important to describe patterns of altered lung structure in COPD. The classification system proposed and illustrated in this article provides a structured approach to visual and quantitative assessment of COPD. Emphysema is classified as centrilobular (subclassified as trace, mild, moderate, confluent, and advanced destructive emphysema), panlobular, and paraseptal (subclassified as mild or substantial). Additional important visual features include airway wall thickening, inflammatory small airways disease, tracheal abnormalities, interstitial lung abnormalities, pulmonary arterial enlargement, and bronchiectasis.
Collapse
Affiliation(s)
- David A Lynch
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - John H M Austin
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - James C Hogg
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Philippe A Grenier
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Hans-Ulrich Kauczor
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Alexander A Bankier
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - R Graham Barr
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Thomas V Colby
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Jeffrey R Galvin
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Pierre Alain Gevenois
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Harvey O Coxson
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Eric A Hoffman
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - John D Newell
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Massimo Pistolesi
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - Edwin K Silverman
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| | - James D Crapo
- From the Departments of Radiology (D.A.L.) and Medicine (J.D.C.), National Jewish Health, 1400 Jackson St, Denver, CO 80206; Department of Radiology, Columbia University, New York, NY (J.H.M.A.); Department of Pathology, University of British Columbia, Vancouver, BC, Canada (J.C.H.); Department of Radiology, Hôpital Pitié-Salpêtrière, Paris, France (P.A.G.); Department of Diagnostic and Interventional Radiology, University of Heidelberg, Heidelberg, Germany (H.U.K.); Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Mass (A.A.B.); Departments of Medicine and Epidemiology, Columbia University Medical Center, New York, NY (R.G.B.); Department of Pathology, Mayo Clinic Scottsdale, Scottsdale, Ariz (T.V.C.); Department of Chest Imaging, American Institute for Radiologic Pathology, Silver Spring, Md (J.R.G.); Department of Radiology, Hôpital Erasme, Brussels, Belgium (P.A.G.); Department of Radiology, Vancouver General Hospital, Vancouver, BC, Canada (H.C.); Department of Radiology, Division of Physiological Imaging, Carver College of Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa (E.A.H., J.D.N.); Respiratory Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy (M.P.); and Channing Laboratory, Brigham and Women's Hospital, Boston, Mass (E.K.S.)
| |
Collapse
|
328
|
Kilic H, Kokturk N, Sari G, Cakır M. Do females behave differently in COPD exacerbation? Int J Chron Obstruct Pulmon Dis 2015; 10:823-30. [PMID: 25977604 PMCID: PMC4418390 DOI: 10.2147/copd.s78952] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION Little is known about whether there is any sex effect on chronic obstructive lung disease (COPD) exacerbations. This study is intended to describe the possible sex-associated differences in exacerbation profile in COPD patients. METHODS A total of 384 COPD patients who were hospitalized due to exacerbation were evaluated retrospectively for their demographics and previous and current exacerbation characteristics. RESULTS The study was conducted on 109 (28%) female patients and 275 (72%) male patients. The mean age was 68.30±10.46 years. Although females had better forced expiratory volume in 1 second and near-normal forced vital capacity, they had much impaired arterial blood gas levels (partial oxygen pressure [PO2] was 36.28 mmHg vs 57.93 mmHg; partial carbon dioxide pressure [PCO2] was 45.97 mmHg vs 42.49 mmHg; P=0.001), indicating severe exacerbation with respiratory failure. More females had two exacerbations and two hospitalizations, while more men had one exacerbation and one hospitalization. Low adherence to treatment and pulmonary embolism were more frequent in females. Females had longer time from the onset of symptoms till the admission and longer hospitalization duration than males. Comorbidities were less in number and different in women (P<0.05). Women were undertreated and using more oral corticosteroids. CONCLUSION Current data showed that female COPD patients might be more prone to have severe exacerbations, a higher number of hospitalizations, and prolonged length of stay for hospitalization. They have a different comorbidity profile and might be undertreated for COPD.
Collapse
Affiliation(s)
- Hatice Kilic
- Department of Pulmonary Medicine, Ankara Atatürk Training and Research Hospital, Gazi University, Ankara, Turkey
| | - Nurdan Kokturk
- Department of Pulmonary Medicine, School of Medicine, Gazi University School of Medicine, Gazi University, Ankara, Turkey
| | - Gulcin Sari
- Department of Pulmonary Medicine, Dr. Nafiz Körez Sincan Devlet Hastanesi, Gazi University, Ankara, Turkey
| | - Mustafa Cakır
- Department of Public Health, School of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
329
|
Lim S, Lam DCL, Muttalif AR, Yunus F, Wongtim S, Lan LTT, Shetty V, Chu R, Zheng J, Perng DW, de Guia T. Impact of chronic obstructive pulmonary disease (COPD) in the Asia-Pacific region: the EPIC Asia population-based survey. ASIA PACIFIC FAMILY MEDICINE 2015; 14:4. [PMID: 25937817 PMCID: PMC4416253 DOI: 10.1186/s12930-015-0020-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/18/2015] [Indexed: 05/20/2023]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a clinical syndrome encompassing a group of chronic, progressive, and debilitating respiratory conditions, that are characterized by incompletely reversible airflow limitation. Within the Asia-Pacific region, prevalence estimates have been derived using various protocols and study methods, and there is little data on the impact of COPD exacerbations. This study aimed to provide a comprehensive picture of the current prevalence and burden of COPD in this region. METHODS A population-based survey was conducted in nine Asia-Pacific territories between 01 February 2012 and 16 May 2012. Overall, 112,330 households were screened to identify eligible subjects (aged ≥40 years, with a physician diagnosis of COPD, chronic bronchitis or emphysema, or with identifiable symptoms of chronic bronchitis). Out of a sample of 69,279 individuals aged ≥40 years, 4,289 subjects with COPD were identified. Data were collected via face-to-face interviews or by fixed-line telephone, using a structured questionnaire. A total of 1,841 completed questionnaires were analyzed. RESULTS The overall estimated COPD prevalence was 6.2%, with 19.1% of subjects having severe COPD. In the 12 months prior to the survey, nearly half of all subjects (46%) had experienced exacerbations, and 19% had been hospitalized as a result of their condition. When subjects were asked about the impact of their condition on employment, 23% said their condition kept them from working, and 42% felt that their condition limited their ability to work or their activities. Of those who reported taking prescription drugs, 20% did not know the name of the drugs they were taking. Prescription of oral corticosteroids was common, with 44% of subjects having used these during the previous year to manage their respiratory symptoms; in contrast, inhaler use was low (25%). Only 37% of subjects had taken a lung function test, and the majority (89%) of those tested did not know their test results. CONCLUSIONS Across the Asia-Pacific territories surveyed, the prevalence of COPD is high, indicating a substantial socioeconomic burden. Our findings suggest that there is considerable room for improvement in the management of COPD, and highlight a need to enhance patient and physician education in the region.
Collapse
Affiliation(s)
- Sam Lim
- />Duke-NUS Graduate School of Medicine, Singapore, Singapore
| | | | | | - Faisal Yunus
- />Department of Pulmonology and Respiratory Medicine, Universitas Indonesia (FMUI), Jakarta, Indonesia
| | - Somkiat Wongtim
- />Department of Internal Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Le Thi Tuyet Lan
- />Respiratory Care Center, University Medical Center, Ho Chi Minh City, Vietnam
| | - Vikram Shetty
- />Takeda Pharmaceuticals (Asia-Pacific) Pte. Ltd, Singapore, Singapore
| | - Romeo Chu
- />Takeda Pharmaceuticals (Asia-Pacific) Pte. Ltd, Singapore, Singapore
| | - Jinping Zheng
- />State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Diahn-Warng Perng
- />Chest Department, Taipei Veterans General Hospital and School of Medicine, National Yang Ming University, Taipei, Taiwan
| | - Teresita de Guia
- />Division of Pulmonary and Critical Care Medicine, Philippine Heart Center, Quezon City, Philippines
| |
Collapse
|
330
|
Lee JH, Cho MH, Hersh CP, McDonald MLN, Wells JM, Dransfield MT, Bowler RP, Lynch DA, Lomas DA, Crapo JD, Silverman EK. IREB2 and GALC are associated with pulmonary artery enlargement in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2015; 52:365-76. [PMID: 25101718 DOI: 10.1165/rcmb.2014-0210oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary hypertension is associated with advanced chronic obstructive pulmonary disease (COPD), although pulmonary vascular changes occur early in the course of the disease. Pulmonary artery (PA) enlargement (PAE) measured by computed tomography correlates with pulmonary hypertension and COPD exacerbation frequency. Genome-wide association studies of PAE in subjects with COPD have not been reported. To investigate whether genetic variants are associated with PAE within subjects with COPD, we investigated data from current and former smokers from the COPDGene Study and the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints study. The ratio of the diameter of the PA to the diameter of the aorta (A) was measured using computed tomography. PAE was defined as PA/A greater than 1. A genome-wide association study for COPD with PAE was performed using subjects with COPD without PAE (PA/A ≤ 1) as a control group. A secondary analysis used smokers with normal spirometry as a control group. Genotyping was performed on Illumina platforms. The results were summarized using fixed-effect meta-analysis. Both meta-analyses revealed a genome-wide significant locus on chromosome 15q25.1 in IREB2 (COPD with versus without PAE, rs7181486; odds ratio [OR] = 1.32; P = 2.10 × 10(-8); versus smoking control subjects, rs2009746; OR = 1.42; P = 1.32 × 10(-9)). PAE was also associated with a region on 14q31.3 near the GALC gene (rs7140285; OR = 1.55; P = 3.75 × 10(-8)). Genetic variants near IREB2 and GALC likely contribute to genetic susceptibility to PAE associated with COPD. This study provides evidence for genetic heterogeneity associated with a clinically important COPD vascular subtype.
Collapse
Affiliation(s)
- Jin Hwa Lee
- 1 Channing Division of Network Medicine, and
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
331
|
de-Torres JP, Wilson DO, Sanchez-Salcedo P, Weissfeld JL, Berto J, Campo A, Alcaide AB, García-Granero M, Celli BR, Zulueta JJ. Lung cancer in patients with chronic obstructive pulmonary disease. Development and validation of the COPD Lung Cancer Screening Score. Am J Respir Crit Care Med 2015; 191:285-91. [PMID: 25522175 DOI: 10.1164/rccm.201407-1210oc] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RATIONALE Patients with chronic obstructive pulmonary disease (COPD) are at high risk for lung cancer (LC) and represent a potential target to improve the diagnostic yield of screening programs. OBJECTIVES To develop a predictive score for LC risk for patients with COPD. METHODS The Pamplona International Early Lung Cancer Detection Program (P-IELCAP) and the Pittsburgh Lung Screening Study (PLuSS) databases were analyzed. Only patients with COPD on spirometry were included. By logistic regression we determined which factors were independently associated with LC in PLuSS and developed a COPD LC screening score (COPD-LUCSS) to be validated in P-IELCAP. MEASUREMENTS AND MAIN RESULTS By regression analysis, age greater than 60, body mass index less than 25 kg/m(2), pack-years history greater than 60, and emphysema presence were independently associated with LC diagnosis and integrated into the COPD-LUCSS, which ranges from 0 to 10 points. Two COPD-LUCSS risk categories were proposed: low risk (scores 0-6) and high risk (scores 7-10). In comparison with low-risk patients, in both cohorts LC risk increased 3.5-fold in the high-risk category. CONCLUSIONS The COPD-LUCSS is a good predictor of LC risk in patients with COPD participating in LC screening programs. Validation in two different populations adds strength to the findings.
Collapse
|
332
|
Asai K, Kobayashi A, Makihara Y, Johnson M. Anti-inflammatory effects of salmeterol/fluticasone propionate 50/250 mcg combination therapy in Japanese patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis 2015; 10:803-11. [PMID: 25945045 PMCID: PMC4407765 DOI: 10.2147/copd.s79842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Using sputum neutrophils as the primary measure, and other inflammation biomarkers, this study evaluated the anti-inflammatory effects of the combination salmeterol 50 mcg and fluticasone propionate 250 mcg (SFC 250) in Japanese patients with chronic obstructive pulmonary disease (COPD). PATIENTS AND METHODS Patients were treated in a randomized, double-blind, parallel group, placebo-controlled trial with SFC 250 twice daily (n=26) or placebo (n=26) for 12 weeks. At the start and end of treatment, inflammation biomarkers (sputum and serum), lung function, and health status (COPD Assessment Test [CAT] questionnaire) were measured. RESULTS Although a numerical decrease in differential neutrophil count was observed from baseline, SFC 250 did not significantly reduce sputum neutrophils compared with placebo, nor were there significant changes from baseline in the other biomarkers (sputum or serum), lung function, or CAT, versus placebo. Squamous epithelial cell contamination in some sputum samples rendered them unacceptable for analysis, which reduced the sample size to n=19 (SFC 250) and n=10 (placebo). However, inclusion of contaminated samples did not affect the overall trend of the outcome. Ad hoc bootstrap statistical analysis showed a 27.9% (SFC 250) and 1.3% (placebo) decrease in sputum neutrophils. Sputum IL-8 decreased by 43.2% after SFC 250 but increased by 48.3% with placebo. Responder analyses showed 42% of patients had ≥20% decrease in neutrophils from baseline; and 47% of patients had a ≥200 pg/mL change in sputum IL-8 following SFC 250 versus 20% after placebo; both changes are considered clinically relevant. CONCLUSION This study provides additional information about inflammation in Japanese COPD patients and is the first to study the anti-inflammatory effects of SFC 250 in this context and population. In the primary analysis, SFC 250 did not produce significant changes from baseline in sputum neutrophil levels or other sputum or serum inflammatory markers compared with placebo. Secondary ad hoc statistical analysis showed that SFC 250 reduced the number of sputum neutrophils and IL-8 compared with placebo.
Collapse
Affiliation(s)
- Kazuhisa Asai
- Department of Respiratory Medicine, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | | | - Yukio Makihara
- Medical Affairs Respiratory Department, GlaxoSmithKline, Tokyo, Japan
| | | |
Collapse
|
333
|
Paoletti M, Cestelli L, Bigazzi F, Camiciottoli G, Pistolesi M. Chronic Obstructive Pulmonary Disease: Pulmonary Function and CT Lung Attenuation Do Not Show Linear Correlation. Radiology 2015; 276:571-8. [PMID: 25848902 DOI: 10.1148/radiol.2015141769] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To determine whether the relationship between pulmonary function and computed tomographic (CT) lung attenuation in chronic obstructive pulmonary disease (COPD), which is traditionally described with single univariate and multivariate statistical models, could be more accurately described with a multiple model estimation approach. MATERIALS AND METHODS The study was approved by the local ethics committee. All participants provided written informed consent. The prediction of the percentage area with CT attenuation values less than -950 HU at inspiration (%LAA-950insp) and less than -910 HU at expiration (%LAA-910exp) obtained with single univariate and multivariate models was compared with that obtained with a multiple model estimation approach in 132 patients with COPD. RESULTS At univariate analysis, %LAA-950insp and %LAA-910exp values higher than the mean value of this cohort (19.1% and 22.0%) showed better correlation with percentage of predicted diffusing capacity of lung for carbon monoxide (Dlco%) than with airflow obstruction (forced expiratory volume in 1 second [FEV1]/vital capacity [VC]). Conversely, %LAA-950insp and %LAA-910exp values lower than the mean value were correlated with FEV1/VC but not with Dlco%. Multiple model estimation performed with two multivariate regressions, each selecting the most appropriate functional variables (FEV1/VC for mild parenchymal destruction, Dlco% and functional residual capacity for severe parenchymal destruction), predicted better than single multivariate regression both %LAA-950insp (R(2) = 0.75 vs 0.46) and %LAA-910exp (R(2) = 0.83 vs 0.63). CONCLUSION The relationship between pulmonary function data and CT densitometric changes in COPD varies with the level of lung attenuation impairment. The nonlinear profile of this relationship is accurately predicted with a multiple model estimation approach.
Collapse
Affiliation(s)
- Matteo Paoletti
- From the Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Lucia Cestelli
- From the Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Francesca Bigazzi
- From the Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Gianna Camiciottoli
- From the Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| | - Massimo Pistolesi
- From the Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
334
|
van Koeverden I, Blanc PD, Bowler RP, Arjomandi M. Secondhand Tobacco Smoke and COPD Risk in Smokers: A COPDGene Study Cohort Subgroup Analysis. COPD 2015; 12:182-9. [PMID: 24983136 PMCID: PMC4820340 DOI: 10.3109/15412555.2014.922173] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Exposure to secondhand tobacco smoke (SHS) can be a risk factor for chronic obstructive pulmonary disease (COPD), but its role among relatively heavy smokers with potential co-exposure to workplace vapors, gas, dust, and fumes (VGDF) has not been studied. METHODS To estimate the contribution of SHS exposure to COPD risk, taking into account smoking effects and work-related exposures to VGDF, we quantified SHS based on survey responses for 1400 ever-employed subjects enrolled in the COPDGene study, all current or former smokers with or without COPD. Occupational exposures to VGDF were quantified based on a job exposure matrix. The associations between SHS and COPD were tested in multivariate logistic regression analyses adjusted for age, sex, VGDF exposure, and cumulative smoking. RESULTS AND DISCUSSION Exposures to SHS at work and at home during adulthood were associated with increased COPD risk: odds ratio (OR) = 1.12 (95% confidence interval [CI]: 1.02-1.23; p = 0.01) and OR = 1.09 (95%CI: 1.00-1.18; p = 0.04) per 10 years of exposure adjusted for smoking and other covariates, respectively. In addition, subjects with employment histories likely to entail exposure to VGDF were more likely to have COPD: OR = 1.52 (95%CI: 1.16-1.98; p < 0.01) (adjusted for other covariates). While adult home SHS COPD risk was attenuated among the heaviest smokers within the cohort, workplace SHS and job VGDF risks persisted in that stratum. CONCLUSION Among smokers all with at least 10 pack-years, adult home and work SHS exposures and occupational VGDF exposure are all associated with COPD.
Collapse
Affiliation(s)
- Ian van Koeverden
- Radboud University Medical Center, Nijmegen, the Netherlands
- Pulmonary Research Group, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Paul D. Blanc
- Division of Occupational and Environmental Medicine, Department of Medicine, University of California, San Francisco, CA, USA
- Division of Pulmonary, Critical Care, Allergy and Immunology, and Sleep Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | | | - Mehrdad Arjomandi
- Pulmonary Research Group, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
- Division of Pulmonary, Critical Care, Allergy and Immunology, and Sleep Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
335
|
De Soyza A, Calverley PM. Large trials, new knowledge: the changing face of COPD management. Eur Respir J 2015; 45:1692-703. [DOI: 10.1183/09031936.00179714] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 02/01/2015] [Indexed: 11/05/2022]
Abstract
Large, well-designed, drug-treatment trials have allowed useful advances to be made in the treatment and diagnosis of chronic obstructive pulmonary disease (COPD). The two main clinical trial designs that provide evidence of effectiveness are randomised controlled trials (RCTs) and observational studies. RCTs are generally considered to provide more robust evidence than that obtained from observational studies and can generate informative secondary analyses in addition to the primary research query. In COPD, however, well-designed comparator-controlled RCTs, although successful, have been shown to have some limitations, such as a lack of generalisability. The findings of observational studies, whilst prone to bias, can generate valuable data and have also provided useful information relating to the efficacy of treatments in the current COPD management guidelines. This review focuses on major COPD studies published since 2007 (including UPLIFT, TIOSPIR, ECLIPSE and COPDGene), and assesses the influence such RCTs and large observational studies have had on our knowledge of COPD, and how these may impact future trial designs.
Collapse
|
336
|
Wilke S, Jones PW, Müllerova H, Vestbo J, Tal-Singer R, Franssen FME, Agusti A, Bakke P, Calverley PM, Coxson HO, Crim C, Edwards LD, Lomas DA, MacNee W, Rennard SI, Yates JC, Wouters EFM, Spruit MA. One-year change in health status and subsequent outcomes in COPD. Thorax 2015; 70:420-5. [PMID: 25782757 DOI: 10.1136/thoraxjnl-2014-205697] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 02/21/2015] [Indexed: 11/04/2022]
Abstract
BACKGROUND Poor health status has been associated with morbidity and mortality in patients with COPD. To date, the impact of changes in health status on these outcomes remains unknown. AIMS To explore the relationship of clinically relevant changes in health status with exacerbation, hospitalisation or death in patients with COPD. METHODS Characteristics and health status (St George's Respiratory Questionnaire, SGRQ) were assessed over a period of 3 years in 2138 patients with COPD enrolled in the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) study: a longitudinal, prospective, observational study. Associations between change in health status (=4 units in SGRQ score) during year 1 and time to first exacerbation, hospitalisation and death during 2-year follow-up were assessed using Kaplan-Meier plots and log-rank test. RESULTS 1832 (85.7%) patients (age 63.4±7.0 years, 65.4% male, FEV1 48.7±15.6% predicted) underwent assessment at baseline and 1 year. Compared with those who deteriorated, patients with improved or stable health status in year 1 have a lower likelihood of exacerbation (HR 0.78 (95% CI 0.67 to 0.89), p<0.001 and 0.84 (0.73 to 0.97), p=0.016, respectively), hospitalisation (0.72 (0.58 to 0.90), p=0.004 and 0.77 (0.62 to 0.96), p=0.023, respectively) or dying (0.61 (0.39 to 0.95), p=0.027 and 0.58 (0.37 to 0.92), p=0.019, respectively) during 2-year follow-up. This effect persisted after stratification for age and the number of exacerbations and hospitalisations during the first year of the study. CONCLUSIONS Patients with stable or improved health status during year 1 of ECLIPSE had a lower likelihood of exacerbation, hospitalisation or dying during 2-year follow-up. Interventions that stabilise and improve health status may also improve outcomes in patients with COPD. TRIAL REGISTRATION NUMBER NCT00292552, registered at ClinicalTrials.gov.
Collapse
Affiliation(s)
- Sarah Wilke
- Department of Research & Education, CIRO+, Centre of Expertise for Chronic Organ Failure, Horn, The Netherlands
| | - Paul W Jones
- Division of Clinical Science, St George's University of London, London, UK
| | - H Müllerova
- Respiratory Epidemiology, GlaxoSmithKline, Uxbridge, UK
| | - Jørgen Vestbo
- Department of Respiratory Medicine, Gentofte Hospital Hellerup, Gentofte, Denmark Research Group, Manchester Academic Health Sciences Centre, University Hospital South Manchester NHS Foundation, Manchester, UK
| | - Ruth Tal-Singer
- Research and Development, GlaxoSmithKline, King of Prussia, UK
| | - Frits M E Franssen
- Department of Research & Education, CIRO+, Centre of Expertise for Chronic Organ Failure, Horn, The Netherlands
| | - Alvar Agusti
- Thorax Institute, Hospital Clinic, IDIBAPS, Universitat de Barcelona and CIBER Enfermedades Respiratorias (CIBERES), Barcelona, Spain
| | - Per Bakke
- Department of Thoracic Medicine, Institute of Clinical Science, University of Bergen, Haukeland University Hospital, Bergen, Norway
| | - Peter M Calverley
- Division of Infection and Immunity Clinical Sciences Centre, University Hospital Aintree, Liverpool, UK
| | - Harvey O Coxson
- Department of Radiology, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - Courtney Crim
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Lisa D Edwards
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - David A Lomas
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - William MacNee
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edingburgh, UK
| | - Stephen I Rennard
- Division of Pulmonary, Critical Care, Sleep & Allergy, University of Nebraska Medical Center, Omaha, USA
| | - Julie C Yates
- GlaxoSmithKline, Research Triangle Park, North Carolina, USA
| | - Emiel F M Wouters
- Department of Research & Education, CIRO+, Centre of Expertise for Chronic Organ Failure, Horn, The Netherlands Department of Respiratory Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martijn A Spruit
- Department of Research & Education, CIRO+, Centre of Expertise for Chronic Organ Failure, Horn, The Netherlands Faculty of Medicine and Life Sciences, REVAL-Rehabilitation Research Center, BIOMED-Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
337
|
Capaldi DP, Sheikh K, Guo F, Svenningsen S, Etemad-Rezai R, Coxson HO, Leipsic JA, McCormack DG, Parraga G. Free-breathing pulmonary 1H and Hyperpolarized 3He MRI: comparison in COPD and bronchiectasis. Acad Radiol 2015; 22:320-9. [PMID: 25491735 DOI: 10.1016/j.acra.2014.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 10/24/2022]
Abstract
RATIONALE AND OBJECTIVES In this proof-of-concept demonstration, we aimed to quantitatively and qualitatively compare pulmonary ventilation abnormalities derived from Fourier decomposition of free-breathing (1)H magnetic resonance imaging (FDMRI) to hyperpolarized (3)He MRI in subjects with chronic obstructive pulmonary disease (COPD) and bronchiectasis. MATERIALS AND METHODS All subjects provided written informed consent to a protocol approved by a local research ethics board and Health, Canada, and they underwent MRI, computed tomography (CT), spirometry, and plethysmography during a single 2-hour visit. Semiautomated segmentation was used to generate ventilation defect measurements derived from FDMRI and (3)He MRI, and these were compared using analysis of variance and Pearson correlations. RESULTS Twenty-six subjects were evaluated including 12 COPD subjects (67 ± 9 years) and 14 bronchiectasis subjects (70 ± 11 years). For COPD subjects, FDMRI and (3)He MRI ventilation defect percent (VDP) was 7 ± 6% and 24 ± 14%, respectively (P < .001; bias = -16 ± 9%). In COPD subjects, FDMRI was significantly correlated with (3)He MRI VDP (r = .88; P = .0001), (3)He MRI apparent diffusion coefficient (r = .71; P < .05), airways resistance (r = .60; P < .05), and RA950 (r = .80; P < .01). In subjects with bronchiectasis, FDMRI VDP (5 ± 3%) and (3)He MRI VDP (18 ± 9%) were significantly different (P < .001) and not correlated (P > .05). The Dice similarity coefficient (DSC) for FDMRI and (3)He MRI ventilation was 86 ± 7% for COPD and 86 ± 4% for bronchiectasis subjects (P > .05); the DSC for FDMRI ventilation defects and CT RA950 was 19 ± 20% in COPD and 2 ± 3% in bronchiectasis subjects (P < .01). CONCLUSIONS FDMRI and (3)He MRI VDP were strongly related in COPD but not in bronchiectasis subjects. In COPD only, FDMRI ventilation defects were spatially related with (3)He ventilation defects and emphysema.
Collapse
|
338
|
Ross CL, Galloway-Phillipps N, Armstrong PC, Mitchell JA, Warner TD, Brearley C, Ito M, Tunstall T, Elkin S, Kon OM, Hansel TT, Paul-Clark MJ. Protocol for a human in vivo model of acute cigarette smoke inhalation challenge in smokers with COPD: monitoring the nasal and systemic immune response using a network biology approach. BMJ Open 2015; 5:e005750. [PMID: 25631307 PMCID: PMC4316420 DOI: 10.1136/bmjopen-2014-005750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Cigarette smoke contributes to a diverse range of diseases including chronic obstructive pulmonary disease (COPD), cardiovascular disorders and many cancers. There currently is a need for human challenge models, to assess the acute effects of a controlled cigarette smoke stimulus, followed by serial sampling of blood and respiratory tissue for advanced molecular profiling. We employ precision sampling of nasal mucosal lining fluid by absorption to permit soluble mediators measurement in eluates. Serial nasal curettage was used for transcriptomic analysis of mucosal tissue. METHODS AND ANALYSIS Three groups of strictly defined patients will be studied: 12 smokers with COPD (GOLD Stage 2) with emphysema, 12 matched smokers with normal lung function and no evidence of emphysema, and 12 matched never smokers with normal spirometry. Patients in the smoking groups are current smokers, and will be given full support to stop smoking immediately after this study. In giving a controlled cigarette smoke stimulus, all patients will have abstained from smoking for 12 h, and will smoke two cigarettes with expiration through the nose in a ventilated chamber. Before and after inhalation of cigarette smoke, a series of samples will be taken from the blood, nasal mucosal lining fluid and nasal tissue by curettage. Analysis of plasma nicotine and metabolites in relation to levels of soluble inflammatory mediators in nasal lining fluid and blood, as well as assessing nasal transcriptomics, ex vivo blood platelet aggregation and leucocyte responses to toll-like receptor agonists will be undertaken. IMPLICATIONS Development of acute cigarette smoke challenge models has promise for the study of molecular effects of smoking in a range of pathological processes. ETHICS AND DISSEMINATION This study was approved by the West London National Research Ethics Committee (12/LO/1101). The study findings will be presented at conferences and will be reported in peer-reviewed journals.
Collapse
Affiliation(s)
- Clare L Ross
- Imperial Clinical Respiratory Research Unit (ICRRU) and Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), St Mary's Hospital, Imperial College, London, UK
| | | | - Paul C Armstrong
- William Harvey Research Institute, Barts and The London, Queen Mary's School of Medicine and Dentistry, London, UK
| | - Jane A Mitchell
- National Heart and Lung Institute, Imperial College, London, UK
| | - Timothy D Warner
- William Harvey Research Institute, Barts and The London, Queen Mary's School of Medicine and Dentistry, London, UK
| | | | - Mari Ito
- Dainippon Sumitomo Pharma Co Ltd, Osaka, Japan
- Department of Molecular Regulation for intractable Diseases, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Tanushree Tunstall
- Imperial Clinical Respiratory Research Unit (ICRRU) and Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), St Mary's Hospital, Imperial College, London, UK
| | - Sarah Elkin
- Imperial Clinical Respiratory Research Unit (ICRRU) and Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), St Mary's Hospital, Imperial College, London, UK
| | - Onn Min Kon
- Imperial Clinical Respiratory Research Unit (ICRRU) and Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), St Mary's Hospital, Imperial College, London, UK
| | - Trevor T Hansel
- Imperial Clinical Respiratory Research Unit (ICRRU) and Biomedical Research Centre (BMRC), Centre for Respiratory Infection (CRI), St Mary's Hospital, Imperial College, London, UK
| | | |
Collapse
|
339
|
Lopez-Campos JL, Calero C, Lopez-Ramirez C, Asensio-Cruz MI, Márquez-Martín E, Ortega-Ruiz F. Patient-reported outcomes and considerations in the management of COPD: focus on aclidinium. Patient Prefer Adherence 2015; 9:95-104. [PMID: 25653503 PMCID: PMC4303404 DOI: 10.2147/ppa.s55009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a complex heterogeneous disease, in which several factors combine to give the final clinical expression. Both early and more recent studies have shown that forced expiratory volume in one second (FEV1), despite being an extremely important parameter to predict the progression of the disease, is a poor surrogate marker for symptoms perception. Accordingly, patient-reported outcomes (PROs) have gained popularity as a measure of the impact of treatment from the patients' perspective, since they represent the individuals' perception of their health status, beyond any physiological limitations. Several such PROs, therefore, are currently included in multidimensional COPD evaluation. This multidimensional approach helps identify different patient types and individualize, up to a certain point, pharmacological treatment. In this multidimensional approach it is important to highlight the importance of long-acting bronchodilators in COPD treatment strategies. Long-acting bronchodilators are cost-effective and have been shown to achieve the greatest functional and clinical improvements in COPD. As a result, long-acting bronchodilators are now the main pharmacological treatment for COPD at all stages of the disease. Until recently, tiotropium was the leading bronchodilator for the treatment of COPD. The clinical development of this medication, unprecedented in inhaled therapy, involved tens of thousands of patients and yielded consistent outcomes in terms of lung function, symptoms, quality of life, exacerbations, and prognosis. However, new long-acting bronchodilators have recently been developed or are currently under development. In this review, we evaluate the effects of aclidinium bromide, a novel long-acting bronchodilator, on PROs in COPD. Aclidinium is a novel long-acting muscarinic antagonist with a good safety profile for the treatment of COPD, and has proven efficacy in both objective functional measurements and PROs. Comparison studies with tiotropium have shown it to have similar lung function improvement and a similar impact on PROs, including quality of life or symptom perception.
Collapse
Affiliation(s)
- Jose Luis Lopez-Campos
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Calero
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Cecilia Lopez-Ramirez
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Maria Isabel Asensio-Cruz
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Eduardo Márquez-Martín
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Francisco Ortega-Ruiz
- Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
340
|
Kardos P, Vogelmeier C, Buhl R, Criée CP, Worth H. The Prospective Non-Interventional DACCORD Study in the National COPD Registry in Germany: design and methods. BMC Pulm Med 2015; 15:2. [PMID: 25578330 PMCID: PMC4417326 DOI: 10.1186/1471-2466-15-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 01/06/2015] [Indexed: 11/24/2022] Open
Abstract
Background A variety of large randomized controlled trials (RCT’s) evaluating pharmacotherapy in chronic obstructive pulmonary disease (COPD) patients does exist. One of the drugs that has been tested is the new long-acting anticholinergic glycopyrronium bromide. Methods As the generalizability of results from RCT’s is questionable we designed a longitudinal, prospective non-interventional study (DACCORD) of two years duration plus two years extension with at least 6000 participants in approximately 500 primary and secondary care practices in Germany (within the new established COPD National Prospective Registry), to assess patient reported outcomes (PRO’s), lung function, adherence and drug safety. To circumvent the hurdle of inappropriate COPD diagnosis in a non-interventional trial, patients have to fulfill the inclusion criteria of the COPD disease management program (DMP) of the German statutory health insurances. Patient management should follow the German national COPD guidelines, which are based on Global Initiative for Chronic Obstructive Lung Disease 2007 (GOLD) report. Labels of prescribed drugs should also be taken into account. Patients received treatment as part of their standard care: at the discretion of the investigator patients were included in one of two arms. A: standard care with glycopyrronium containing regimen, and arm B: standard care without glycopyrronium. Discussion For 2016 we expect important results regarding longitudinal development of PRO’s including exacerbations, lung function, adherence and side effects. We also investigate applicability of the new GOLD staging system in usual care. Data on diagnostic and treatment modalities in current German primary and secondary care, as well as pharmaco-economic data will be generated. Trial registration 1. German Register for non-interventional studies: http://www.vfa.de/de/arzneimittel-forschung/datenbanken-zu-arzneimitteln/nisdb. 2. EMA EnCePP http://www.encepp.eu/.
Collapse
Affiliation(s)
- Peter Kardos
- Group Practice and Centre for Allergy, Respiratory and Sleep Medicine, Red Cross Maingau Hospital, Scheffelstrasse 33, 60318, Frankfurt am Main, Germany.
| | - Claus Vogelmeier
- Department of Respiratory Diseases, University of Marburg, 35043, Marburg, Germany.
| | - Roland Buhl
- Pulmonary Department, Mainz University Hospital, 55131, Mainz, Germany.
| | - Carl-Peter Criée
- Department of Sleep and Respiratory Medicine, Evangelical Hospital Goettingen-Weende, 37120, Bovenden, Germany.
| | - Heinrich Worth
- Department of Pulmonology and Cardiology, Hospital Fuerth, University Erlangen-Nuernberg, 90766, Fuerth, Germany.
| |
Collapse
|
341
|
McDonald MLN, Cho MH, Sørheim IC, Lutz SM, Castaldi PJ, Lomas DA, Coxson HO, Edwards LD, MacNee W, Vestbo J, Yates JC, Agusti A, Calverley PMA, Celli B, Crim C, Rennard SI, Wouters EFM, Bakke P, Tal-Singer R, Miller BE, Gulsvik A, Casaburi R, Wells JM, Regan EA, Make BJ, Hokanson JE, Lange C, Crapo JD, Beaty TH, Silverman EK, Hersh CP. Common genetic variants associated with resting oxygenation in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2015; 51:678-87. [PMID: 24825563 DOI: 10.1165/rcmb.2014-0135oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Hypoxemia is a major complication of chronic obstructive pulmonary disease (COPD) that correlates with disease prognosis. Identifying genetic variants associated with oxygenation may provide clues for deciphering the heterogeneity in prognosis among patients with COPD. However, previous genetic studies have been restricted to investigating COPD candidate genes for association with hypoxemia. To report results from the first genome-wide association study (GWAS) of resting oxygen saturation (as measured by pulse oximetry [Spo2]) in subjects with COPD, we performed a GWAS of Spo2 in two large, well characterized COPD populations: COPDGene, including both the non-Hispanic white (NHW) and African American (AA) groups, and Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE). We identified several suggestive loci (P < 1 × 10(-5)) associated with Spo2 in COPDGene in the NHW (n = 2810) and ECLIPSE (n = 1758) groups, and two loci on chromosomes 14 and 15 in the AA group (n = 820) from COPDGene achieving a level of genome-wide significance (P < 5 × 10(-8)). The chromosome 14 single-nucleotide polymorphism, rs6576132, located in an intergenic region, was nominally replicated (P < 0.05) in the NHW group from COPDGene. The chromosome 15 single-nucleotide polymorphisms were rare in subjects of European ancestry, so the results could not be replicated. The chromosome 15 region contains several genes, including TICRR and KIF7, and is proximal to RHCG (Rh family C glyocoprotein gene). We have identified two loci associated with resting oxygen saturation in AA subjects with COPD, and several suggestive regions in subjects of European descent with COPD. Our study highlights the importance of investigating the genetics of complex traits in different racial groups.
Collapse
|
342
|
Dijkstra AE, Boezen HM, van den Berge M, Vonk JM, Hiemstra PS, Barr RG, Burkart KM, Manichaikul A, Pottinger TD, Silverman EK, Cho MH, Crapo JD, Beaty TH, Bakke P, Gulsvik A, Lomas DA, Bossé Y, Nickle DC, Paré PD, de Koning HJ, Lammers JW, Zanen P, Smolonska J, Wijmenga C, Brandsma CA, Groen HJM, Postma DS. Dissecting the genetics of chronic mucus hypersecretion in smokers with and without COPD. Eur Respir J 2015; 45:60-75. [PMID: 25234806 PMCID: PMC4498483 DOI: 10.1183/09031936.00093314] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Smoking is a notorious risk factor for chronic mucus hypersecretion (CMH). CMH frequently occurs in chronic obstructive pulmonary disease (COPD). The question arises whether the same single-nucleotide polymorphisms (SNPs) are related to CMH in smokers with and without COPD. We performed two genome-wide association studies of CMH under an additive genetic model in male heavy smokers (≥20 pack-years) with COPD (n=849, 39.9% CMH) and without COPD (n=1348, 25.4% CMH), followed by replication and meta-analysis in comparable populations, and assessment of the functional relevance of significantly associated SNPs. Genome-wide association analysis of CMH in COPD and non-COPD subjects yielded no genome-wide significance after replication. In COPD, our top SNP (rs10461985, p=5.43×10(-5)) was located in the GDNF-AS1 gene that is functionally associated with the GDNF gene. Expression of GDNF in bronchial biopsies of COPD patients was significantly associated with CMH (p=0.007). In non-COPD subjects, four SNPs had a p-value <10(-5) in the meta-analysis, including a SNP (rs4863687) in the MAML3 gene, the T-allele showing modest association with CMH (p=7.57×10(-6), OR 1.48) and with significantly increased MAML3 expression in lung tissue (p=2.59×10(-12)). Our data suggest the potential for differential genetic backgrounds of CMH in individuals with and without COPD.
Collapse
Affiliation(s)
- Akkelies E Dijkstra
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - H Marike Boezen
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Maarten van den Berge
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Judith M Vonk
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Pieter S Hiemstra
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - R Graham Barr
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Kirsten M Burkart
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Ani Manichaikul
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Tess D Pottinger
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Edward K Silverman
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Michael H Cho
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - James D Crapo
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Terri H Beaty
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Per Bakke
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Amund Gulsvik
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - David A Lomas
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Yohan Bossé
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - David C Nickle
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Peter D Paré
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Harry J de Koning
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Jan-Willem Lammers
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Pieter Zanen
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Joanna Smolonska
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Ciska Wijmenga
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Corry-Anke Brandsma
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Harry J M Groen
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| | - Dirkje S Postma
- For lists of the authors' affiliations, and the LifeLines Cohort Study group members and their affiliations, see the Acknowledgements section
| |
Collapse
|
343
|
Kirby M, Pike D, Coxson HO, McCormack DG, Parraga G. Hyperpolarized3He Ventilation Defects Used to Predict Pulmonary Exacerbations in Mild to Moderate Chronic Obstructive Pulmonary Disease. Radiology 2014; 273:887-96. [DOI: 10.1148/radiol.14140161] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
344
|
Márquez-Martín E, Ruiz FO, Ramos PC, López-Campos JL, Azcona BV, Cortés EB. Randomized trial of non-invasive ventilation combined with exercise training in patients with chronic hypercapnic failure due to chronic obstructive pulmonary disease. Respir Med 2014; 108:1741-51. [DOI: 10.1016/j.rmed.2014.10.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/11/2014] [Accepted: 10/14/2014] [Indexed: 10/24/2022]
|
345
|
Roca J, Vargas C, Cano I, Selivanov V, Barreiro E, Maier D, Falciani F, Wagner P, Cascante M, Garcia-Aymerich J, Kalko S, De Mas I, Tegnér J, Escarrabill J, Agustí A, Gomez-Cabrero D. Chronic Obstructive Pulmonary Disease heterogeneity: challenges for health risk assessment, stratification and management. J Transl Med 2014; 12 Suppl 2:S3. [PMID: 25472887 PMCID: PMC4255905 DOI: 10.1186/1479-5876-12-s2-s3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background and hypothesis Heterogeneity in clinical manifestations and disease progression in Chronic Obstructive Pulmonary Disease (COPD) lead to consequences for patient health risk assessment, stratification and management. Implicit with the classical "spill over" hypothesis is that COPD heterogeneity is driven by the pulmonary events of the disease. Alternatively, we hypothesized that COPD heterogeneities result from the interplay of mechanisms governing three conceptually different phenomena: 1) pulmonary disease, 2) systemic effects of COPD and 3) co-morbidity clustering, each of them with their own dynamics. Objective and method To explore the potential of a systems analysis of COPD heterogeneity focused on skeletal muscle dysfunction and on co-morbidity clustering aiming at generating predictive modeling with impact on patient management. To this end, strategies combining deterministic modeling and network medicine analyses of the Biobridge dataset were used to investigate the mechanisms of skeletal muscle dysfunction. An independent data driven analysis of co-morbidity clustering examining associated genes and pathways was performed using a large dataset (ICD9-CM data from Medicare, 13 million people). Finally, a targeted network analysis using the outcomes of the two approaches (skeletal muscle dysfunction and co-morbidity clustering) explored shared pathways between these phenomena. Results (1) Evidence of abnormal regulation of skeletal muscle bioenergetics and skeletal muscle remodeling showing a significant association with nitroso-redox disequilibrium was observed in COPD; (2) COPD patients presented higher risk for co-morbidity clustering than non-COPD patients increasing with ageing; and, (3) the on-going targeted network analyses suggests shared pathways between skeletal muscle dysfunction and co-morbidity clustering. Conclusions The results indicate the high potential of a systems approach to address COPD heterogeneity. Significant knowledge gaps were identified that are relevant to shape strategies aiming at fostering 4P Medicine for patients with COPD.
Collapse
|
346
|
Nobakht M Gh BF, Aliannejad R, Rezaei-Tavirani M, Taheri S, Oskouie AA. The metabolomics of airway diseases, including COPD, asthma and cystic fibrosis. Biomarkers 2014; 20:5-16. [PMID: 25403491 DOI: 10.3109/1354750x.2014.983167] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD), asthma and cystic fibrosis (CF) are characterized by airway obstruction and an inflammatory process. Reaching early diagnosis and discrimination of subtypes of these respiratory diseases are quite a challenging task than other chronic illnesses. Metabolomics is the study of metabolic pathways and the measurement of unique biochemical molecules generated in a living system. In the last decade, metabolomics has already proved to be useful for the characterization of several pathological conditions and offers promises as a clinical tool. In this article, we review the current state of the metabolomics of COPD, asthma and CF with a focus on the different methods and instrumentation being used for the discovery of biomarkers in research and translation into clinic as diagnostic aids for the choice of patient-specific therapies.
Collapse
Affiliation(s)
- B Fatemeh Nobakht M Gh
- Faculty of Paramedical Sciences, Proteomics Research Center, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | | | | | | | | |
Collapse
|
347
|
Aarli BB, Calverley PM, Jensen RL, Eagan TM, Bakke PS, Hardie JA. Variability of within-breath reactance in COPD patients and its association with dyspnoea. Eur Respir J 2014; 45:625-34. [DOI: 10.1183/09031936.00051214] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The forced oscillation technique can identify expiratory flow limitation (EFL) when a large difference in inspiratory and expiratory reactance (ΔXrs) occurs. However, flow limitation can vary from breath to breath, and so we compared a multiple-breath ΔXrs approach to the traditional breath-by-breath assessment of EFL. We investigated the within- and between-day reproducibility and the factors that affect the size of ΔXrs when used as a continuous measurement over multiple breaths. In addition, we examined how multiple-breath ΔXrs relates to the sensation of breathlessness.425 moderate to very severe chronic obstructive pulmonary disease (COPD) patients and 229 controls were included. Spirometry and impedance measurements were performed on a MasterScope CT Impulse Oscillation System.Median ΔXrs approached zero in healthy controls with little variation between measurements. COPD patients generally had higher ΔXrs and higher variability. The COPD patients with ΔXrs >0.1 kPa·L−1·s−1 were prone to be more breathless and had a higher modified Medical Research Council dyspnoea scale score. In controls, the 95th percentile of ΔXrs was as low as 0.07 kPa·L−1·s−1.We describe a new method to assess EFL at a patient level and propose a cut-off, mean ΔXrs >0.1 kPa·L−1·s−1, as a way to identify COPD patients who are more likely to report dyspnoea.
Collapse
|
348
|
Hersh CP, Make BJ, Lynch DA, Barr RG, Bowler RP, Calverley PMA, Castaldi PJ, Cho MH, Coxson HO, DeMeo DL, Foreman MG, Han MK, Harshfield BJ, Hokanson JE, Lutz S, Ramsdell JW, Regan EA, Rennard SI, Schroeder JD, Sciurba FC, Steiner RM, Tal-Singer R, van Beek EJR, Silverman EK, Crapo JD. Non-emphysematous chronic obstructive pulmonary disease is associated with diabetes mellitus. BMC Pulm Med 2014; 14:164. [PMID: 25341556 PMCID: PMC4216374 DOI: 10.1186/1471-2466-14-164] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 10/09/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) has been classically divided into blue bloaters and pink puffers. The utility of these clinical subtypes is unclear. However, the broader distinction between airway-predominant and emphysema-predominant COPD may be clinically relevant. The objective was to define clinical features of emphysema-predominant and non-emphysematous COPD patients. METHODS Current and former smokers from the Genetic Epidemiology of COPD Study (COPDGene) had chest computed tomography (CT) scans with quantitative image analysis. Emphysema-predominant COPD was defined by low attenuation area at -950 Hounsfield Units (LAA-950) ≥10%. Non-emphysematous COPD was defined by airflow obstruction with minimal to no emphysema (LAA-950 < 5%). RESULTS Out of 4197 COPD subjects, 1687 were classified as emphysema-predominant and 1817 as non-emphysematous; 693 had LAA-950 between 5-10% and were not categorized. Subjects with emphysema-predominant COPD were older (65.6 vs 60.6 years, p < 0.0001) with more severe COPD based on airflow obstruction (FEV1 44.5 vs 68.4%, p < 0.0001), greater exercise limitation (6-minute walk distance 1138 vs 1331 ft, p < 0.0001) and reduced quality of life (St. George's Respiratory Questionnaire score 43 vs 31, p < 0.0001). Self-reported diabetes was more frequent in non-emphysematous COPD (OR 2.13, p < 0.001), which was also confirmed using a strict definition of diabetes based on medication use. The association between diabetes and non-emphysematous COPD was replicated in the ECLIPSE study. CONCLUSIONS Non-emphysematous COPD, defined by airflow obstruction with a paucity of emphysema on chest CT scan, is associated with an increased risk of diabetes. COPD patients without emphysema may warrant closer monitoring for diabetes, hypertension, and hyperlipidemia and vice versa. TRIAL REGISTRATION Clinicaltrials.gov identifiers: COPDGene NCT00608764, ECLIPSE NCT00292552.
Collapse
Affiliation(s)
- Craig P Hersh
- />Channing Division of Network Medicine, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Barry J Make
- />Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, CO USA
| | - David A Lynch
- />Department of Radiology, National Jewish Health, Denver, CO USA
| | - R Graham Barr
- />Department of Medicine, Columbia University, New York, NY USA
| | - Russell P Bowler
- />Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, CO USA
| | - Peter MA Calverley
- />Division of Infection and Immunity Clinical Sciences Centre, University Hospital Aintree, Liverpool, UK
| | | | - Michael H Cho
- />Channing Division of Network Medicine, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Harvey O Coxson
- />Department of Radiology, University of British Columbia, Vancouver, Canada
| | - Dawn L DeMeo
- />Channing Division of Network Medicine, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - Marilyn G Foreman
- />Division of Pulmonary and Critical Care Medicine, Morehouse School of Medicine, Atlanta, GA USA
| | - MeiLan K Han
- />Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, MI USA
| | | | - John E Hokanson
- />Department of Epidemiology, Colorado School of Public Health, Aurora, CO USA
| | - Sharon Lutz
- />Department of Biostatistics, Colorado School of Public Health, Aurora, CO USA
| | - Joe W Ramsdell
- />Division of Pulmonary and Critical Care Medicine, University of California, San Diego, CA USA
| | - Elizabeth A Regan
- />Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, CO USA
| | - Stephen I Rennard
- />Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE USA
| | | | - Frank C Sciurba
- />Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Robert M Steiner
- />Department of Radiology, Temple University, Philadelphia, PA USA
| | | | - Edwin JR van Beek
- />Department of Radiology, University of Edinburgh, Edinburgh, Scotland
| | - Edwin K Silverman
- />Channing Division of Network Medicine, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - James D Crapo
- />Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, CO USA
| | - the COPDGene and ECLIPSE Investigators
- />Channing Division of Network Medicine, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
- />Division of Pulmonary and Critical Care Medicine, National Jewish Health, Denver, CO USA
- />Department of Radiology, National Jewish Health, Denver, CO USA
- />Department of Medicine, Columbia University, New York, NY USA
- />Division of Infection and Immunity Clinical Sciences Centre, University Hospital Aintree, Liverpool, UK
- />Department of Radiology, University of British Columbia, Vancouver, Canada
- />Division of Pulmonary and Critical Care Medicine, Morehouse School of Medicine, Atlanta, GA USA
- />Division of Pulmonary and Critical Care Medicine, University of Michigan Health System, Ann Arbor, MI USA
- />Department of Epidemiology, Colorado School of Public Health, Aurora, CO USA
- />Department of Biostatistics, Colorado School of Public Health, Aurora, CO USA
- />Division of Pulmonary and Critical Care Medicine, University of California, San Diego, CA USA
- />Division of Pulmonary, Critical Care, Sleep and Allergy, University of Nebraska Medical Center, Omaha, NE USA
- />Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA USA
- />Department of Radiology, Temple University, Philadelphia, PA USA
- />GlaxoSmithKline R&D, King of Prussia, PA USA
- />Department of Radiology, University of Edinburgh, Edinburgh, Scotland
| |
Collapse
|
349
|
Singh D, Kolsum U, Brightling CE, Locantore N, Agusti A, Tal-Singer R. Eosinophilic inflammation in COPD: prevalence and clinical characteristics. Eur Respir J 2014; 44:1697-700. [PMID: 25323230 DOI: 10.1183/09031936.00162414] [Citation(s) in RCA: 339] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Dave Singh
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, Manchester, UK
| | - Umme Kolsum
- University of Manchester, Medicines Evaluation Unit, University Hospital of South Manchester Foundation Trust, Manchester, UK
| | - Chris E Brightling
- Infection, Immunity and Inflammation, University of Leicester, Leicester, UK
| | - Nicholas Locantore
- GlaxoSmithKline, Research and Development, Research Triangle Park, NC, USA
| | - Alvar Agusti
- Fundació Investigació Sanitària Illes Balears (FISIB), Centro de investigación biomedical en red de enfermedades respiratorias (CIBERES), Palma de Mallorca, Spain
| | - Ruth Tal-Singer
- GlaxoSmithKline, Research and Development, King of Prussia, PA, USA
| | | |
Collapse
|
350
|
Diaz AA, Zhou L, Young TP, McDonald ML, Harmouche R, Ross JC, San Jose Estepar R, Wouters EFM, Coxson HO, MacNee W, Rennard S, Maltais F, Kinney GL, Hokanson JE, Washko GR. Chest CT measures of muscle and adipose tissue in COPD: gender-based differences in content and in relationships with blood biomarkers. Acad Radiol 2014; 21:1255-61. [PMID: 25088837 DOI: 10.1016/j.acra.2014.05.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/01/2014] [Accepted: 05/03/2014] [Indexed: 11/17/2022]
Abstract
RATIONALE AND OBJECTIVES Computed tomography (CT) of the chest can be used to assess pectoralis muscle area (PMA) and subcutaneous adipose tissue (SAT) area. Adipose tissue content is associated with inflammatory mediators in chronic obstructive pulmonary disease (COPD) subjects. Based on gender differences in body composition, we aimed to assess the hypothesis that in subjects with COPD, the relationships between PMA, SAT, and blood biomarkers of inflammation differ by gender. MATERIALS AND METHODS We compared chest CT measures of PMA and SAT on a single slice at aortic arch and supraesternal notch levels from 73 subjects (28 women) with COPD between genders. The relationships of PMA and SAT area to biomarkers were assessed using within-gender regression models. RESULTS Women had a lesser PMA and a greater SAT area than men (difference range for PMA, 13.3-22.8 cm²; for SAT, 11.8-12.4 cm²; P < .05 for all comparisons) at both anatomic levels. These differences in PMA and SAT remained significant after adjustment for age and body mass index. Within-gender regression models adjusted for age showed that SAT was directly associated with C-reactive protein (for aortic arch level, P = .04) and fibrinogen (for both anatomic locations, P = .003) only in women, whereas PMA was not associated with any biomarkers in either gender. CONCLUSIONS It appears that in subjects with COPD, there are gender-based differences in the relationships between subcutaneous adipose tissue and inflammatory biomarkers.
Collapse
Affiliation(s)
- Alejandro A Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115.
| | - Linfu Zhou
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115
| | - Tom P Young
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115
| | - Merry-Lynn McDonald
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rola Harmouche
- Surgical Planning Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - James C Ross
- Surgical Planning Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raul San Jose Estepar
- Surgical Planning Laboratory, Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Harvey O Coxson
- Department of Radiology and James Hogg Research Centre, Institute for Heart and Lung Health, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | - François Maltais
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Quebec, Canada
| | - Gregory L Kinney
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado, Aurora, Colorado
| | - George R Washko
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis St, Boston, MA 02115
| |
Collapse
|