You are on page 1of 128

GOLD_WR_05 8/18/05 12:56 PM Page A1

Global Initiative for Chronic


Obstructive
Lung
Disease

GLOBAL STRATEGY FOR THE DIAGNOSIS,


MANAGEMENT, AND PREVENTION OF
CHRONIC OBSTRUCTIVE PULMONARY DISEASE
UPDATED 2005
GOLD_WR_05 8/18/05 12:56 PM Page i

GLOBAL INITIATIVE FOR


CHRONIC OBSTRUCTIVE LUNG DISEASE
GLOBAL STRATEGY FOR THE DIAGNOSIS, MANAGEMENT,
AND PREVENTION OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE
Updated 2005 (Based on an April 1998 NHLBI/WHO Workshop)

i
GOLD_WR_05 8/18/05 12:56 PM Page ii

APRIL 1998 WORKSHOP PANEL


Global Strategy for the Diagnosis, Management, and Prevention of
Chronic Obstructive Pulmonary Disease: NHLBI/WHO Workshop
National Heart, Lung, and Blood Institute: Claude Lenfant, MD
World Health Organization: Nikolai Khaltaev, MD

Romain Pauwels, MD, PhD, Chair Christine Jenkins, MD


Ghent University Hospital Concord Hospital
Ghent, Belgium Sydney, New South Wales, Australia

Nicholas Anthonisen, MD Dirkje S. Postma, MD


University of Manitoba Academic Hospital Groningen
Winnipeg, Manitoba, Canada Groningen, the Netherlands

William C. Bailey, MD Klaus F. Rabe, MD


University of Alabama at Birmingham Leiden University Medical Center
Birmingham, Alabama, US Leiden, the Netherlands

Peter J. Barnes, MD Scott D. Ramsey, MD, PhD


National Heart & Lung Institute University of Washington
London, UK Seattle, Washington, US

A. Sonia Buist, MD Stephen I. Rennard, MD


Oregon Health Sciences University University of Nebraska Medical Center
Portland, Oregon, US Omaha, Nebraska, US

Peter Calverley, MD Roberto Rodriguez-Roisin, MD


University Hospital, Aintree University of Barcelona
Liverpool, UK Barcelona, Spain

Tim Clark, MD Nikos Siafakas, MD


Imperial College University of Crete Medical School
London, UK Heraklion, Greece

Leonardo Fabbri, MD Sean D. Sullivan, PhD


University of Modena & Reggio Emilia University of Washington
Modena, Italy Seattle, Washington, US

Yoshinosuke Fukuchi, MD Wan-Cheng Tan, MD


Juntendo University National University Hospital
Tokyo, Japan Singapore

Lawrence Grouse, MD, PhD GOLD Staff


University of Washington
Seattle, Washington, US Sarah DeWeerdt
Editor
James C. Hogg, MD Seattle, Washington, US
St. Paul’s Hospital
Vancouver, British Columbia, Canada Suzanne S. Hurd, PhD
Scientific Director
Bethesda, Maryland, US

ii
GOLD_WR_05 8/18/05 12:56 PM Page iii

ONSULTANT REVIEWERS
(FOR 1998 WORKSHOP PANEL REPORT)
Individuals

Sherwood Burge (UK) Australian Lung Foundation


Moira Chan-Yeung (Hong Kong) Robert Edwards
James Donohue (US)
Nicholas J. Gross (US) Belgian Society of Pneumology
Helgo Magnussen (Germany) Marc Decramer
Donald Mahler (US) Jean-Claude Yernault
Jean-Francois Muir (France)
Mrigrendra Pandey (India) British Thoracic Society
Peter Pare (Canada) Neil Pride
Thomas Petty (US)
Michael Plit (South Africa) Canadian Thoracic Society
Sri Ram (US) Louis-Philippe Boulet
Harold Rea (New Zealand) Kenneth Chapman
Andrea Rossi (Italy)
Maureen Rutten-van Molken (The Netherlands) Chinese Respiratory Society
Marina Saetta (Italy) Nan-Shan Zhong
Raj Singh (India) Yuanjue Zhu
Frank Speizer (US)
Robert Stockley (UK) Croatian Respiratory Society
Donald Tashkin (US) Neven Rakusic
Ian Town (New Zealand) Davor Plavec
Paul Vermeire (Belgium)
Gregory Wagner (US) Czech Thoracic Society
Scott Weiss (US) Stanislav Kos
Miel Wouters (The Netherlands) Jaromir Musil
Jan Zielinski (Poland) Vladimir Vondra

European Respiratory Society


Organizations Marc Decramer (Belgium)

American College of Chest Physicians French Speaking Pneumological Society


Suzanne Pingleton Michel Fournier
Thomas Similowski
American Thoracic Society
Bart Celli Hungarian Respiratory Society
William Martin Pal Magyar

Austrian Respiratory Society Japanese Respiratory Society


Friedriech Kummer Yoshinosuke Fukuchi

Arab Respiratory Society Latin American Thoracic Society


Salem El Sayed Juan Figueroa (Argentina)
Maria Christina Machado (Brazil)
Thoracic Society of Australia and New Zealand Ilma Paschoal (Brazil)
Alastair Stewart Jose Jardim (Brazil)
David McKenzie Gisele Borzone (Chile)
Peter Frith Orlando Diaz (Chile)

iii
GOLD_WR_05 8/18/05 12:56 PM Page iv

Patricio Gonzales (Chile)


Carmen Lisboa (Chile)
Rogelio Perez Padilla (Mexico)
Jorge Rodriguez De Marco (Uruguay)
Maria Victorina Lopez (Uruguay)
Roberto Lopez (Uruguay)

Malaysian Thoracic Society


Zin Zainudin

Norwegian Thoracic Society


Amund Gulsvik
Ernst Omenaas

Polish Phthisiopneumonological Society


Michal Pirozynski

Romanian Society of Pulmonary Diseases


Traian Mihaescu
Sabina Antoniu

Singapore Thoracic Society


Alan Ng
Wei Keong

Slovakian Pneumological and Phthisiological Society


Ladislav Chovan

Slovenian Respiratory Society


Stanislav Suskovic

South African Thoracic Society


James Joubert

Spanish Society of Pneumology


Teodoro Montemayor Rubio
Victor Sobradillo

Swedish Society for Chest Physicians


Kjell Larsson
Sven Larsson
Claes-Goran Lofdahl

Swiss Pulmonary Society


Philippe Leuenberger
Erich Russi

Thoracic Society of Thailand


Ploysongsang Youngyudh

Vietnam Asthma-Allergology and Clinical


Immunology Association
Nguyen Nang An

iv
GOLD_WR_05 8/18/05 12:56 PM Page v

PREFACE

hronic Obstructive Pulmonary Disease (COPD) is a Development of the Workshop Report was supported
C major public health problem. It is the fourth leading
cause of chronic morbidity and mortality in the United
through educational grants from Altana, Andi-Ventis,
AstraZeneca, Aventis, Bayer, Boehringer Ingelheim,
States1 and is projected to rank fifth in 2020 as a world- Chiesi, GlaxoSmithKline, Merck, Sharp & Dohme,
wide burden of disease according to a study published by Mitsubishi Pharma, Nikken Chemicals, Novartis, Pfizer,
the World Bank/World Health Organization2. Yet, COPD Schering-Plough, and Zambon.
fails to receive adequate attention from the health care
community and government officials. With these con- Leonardo Fabbri, MD
cerns in mind, a committed group of scientists encour- Modena, Italy
aged the US National Heart, Lung, and Blood Institute Chair, GOLD Executive Committee
and the World Health Organization to form the Global
Initiative for Chronic Obstructive Lung Disease (GOLD).
Among GOLD’s important objectives are to increase REFERENCES
awareness of COPD and to help the thousands of people
who suffer from this disease and die prematurely from 1. National Heart, Lung, and Blood Institute. Morbidity &
COPD or its complications. mortality: chartbook on cardiovascular, lung, and blood dis-
eases. Bethesda, MD: US Department of Health and Human
The first step in the GOLD program was to prepare a Services, Public Health Service, National Institutes of Health;
consensus Workshop Report, Global Strategy for the 1998. Available from: URL:
Diagnosis, Management, and Prevention of COPD. The www.nhlbi.nih.gov/nhlbi/seiin//other/cht-book/htm
GOLD Expert Panel, a distinguished group of health 2. Murray CJL, Lopez AD. Evidence-based health
professionals from the fields of respiratory medicine, policy-lessons from the Global Burden of Disease Study.
epidemiology, socioeconomics, public health, and health Science 1996; 274:740-3.
education, reviewed existing COPD guidelines, as well as
new information on pathogenic mechanisms of COPD as
they developed a consensus document. Many recom-
mendations will require additional study and evaluation as
the GOLD program is implemented.

A major problem is the incomplete information about the


causes and prevalence of COPD, especially in developing
countries. While cigarette smoking is a major known risk
factor, much remains to be learned about other causes of
this disease. The GOLD Initiative will bring COPD to the
attention of governments, public health officials, health
care workers, and the general public, but a concerted
effort by all involved in health care will be necessary to
control this major public health problem.

I would like to acknowledge the expert panel that prepared


the first workshop report, and the GOLD Science Committee
for its work in preparation of the yearly updated volumes.
We look forward to our continued work with interested
organizations and the GOLD National Leaders to meet
the goals of the GOLD initiative.

v
GOLD_WR_05 8/18/05 12:56 PM Page vi

Methodology and Summary of New Recommendations (2005 Update)

The GOLD Workshop Report, Global Strategy for GOLD Workshop Report (2005 Update):
Diagnosis, Management and Prevention of COPD Summary of Recommendations
presented in 2001 was based on the scientific literature
available until mid 2000.. To assure that the recommen- Between January 1 and December 2004, 131 articles met
dations for management of COPD remained as current the search criteria. Of these, 10 papers were identified to
as possible, the GOLD Executive Committee established have an impact on the GOLD report. Of these, 5 papers
a Science Committee* to review published research and confirmed an existing statement and were added as a
to post an updated report yearly on the GOLD website. reference:
The first (2003) and second (2004) updates were posted
on the GOLD website (www.gold.copd.org) in July 2003 1. Page 68: Man WD, Mustfa N, Nikoletou D, Kaul S,
and July 2004 respectively. This third update (July 2005) Hart N, Rafferty GF, Donaldson N, Polkey MI, Moxham J.
includes review of publications from January to Effect of salmeterol on respiratory muscle activity during
December 2004. This will be the final update of the 2001 exercise in poorly reversible COPD. Thorax. 2004
document; a revision of the entire document has been Jun;59(6):471-6.
implemented and is scheduled to be completed in 2006.
2. Page 68: O'Donnell DE, Fluge T, Gerken F, Hamilton
Methods: The process used for the 2005 update, identical A, Webb K, Aguilaniu B, Make B, Magnussen H. Effects
to that described for the previous updates, included a of tiotropium on lung hyperinflation, dyspnoea and exercise
Pub Med search using fields established by the tolerance in COPD. Eur Respir J. 2004 Jun;23(6):832-40.
Committee: 1) COPD OR chronic bronchitis OR emphy-
sema, All Fields, All Adult, 19+ years, only items with 3. Page 68: Oostenbrink JB, Rutten-van Molken MP, Al
abstracts, Clinical Trial, Human, sorted by Authors; and MJ, Van Noord JA, Vincken W. One-year cost-effectiveness
2) COPD OR chronic bronchitis OR emphysema AND of tiotropium versus ipratropium to treat chronic obstructive
systematic, All fields, All adult, 19+ years, only items pulmonary disease. Eur Respir J. 2004 Feb;23(2):241-9.
with abstracts, Human, sorted by Author. In addition,
publications in peer review journals not captured by Pub 4. Page 71: Spencer S, Calverley PM, Burge PS, Jones
Med could be submitted to individual members of the PW. Impact of preventing exacerbations on deterioration
Committee providing an abstract and the full paper were of health status in COPD. Eur Respir J. 2004
submitted in (or translated into) English. May;23(5):698-702.

All members of the Committee received a summary of 5. Page 73: Wongsurakiat P, Maranetra KN, Wasi C,
citations and all abstracts. Each abstract was assigned Kositanont U, Dejsomritrutai W, Charoenratanakul S.
to 2 Committee members (members were not assigned Acute respiratory illness in patients with COPD and the
to a paper where he/she appears as an author), although effectiveness of influenza vaccination: a randomized
any member was offered the opportunity to provide an controlled study. Chest. 2004 Jun;125(6):2011-20.
opinion on any abstract. Members evaluated the abstract
or, up to her/his judgment, the full publication, by answering Five papers introduced information that required a new
specific written questions from a short questionnaire, and statement to be added to the report:
to indicate if the scientific data presented impacted on
recommendations in the GOLD report. If so, the member 1. Page 67 – Add sentence: In a study of mild to moderate
was asked to specifically identify modifications that COPD patients at an out-patient clinic, patient education
should be made. The GOLD Science Committee met involving one four hour group session followed by one to
on a regular basis to discuss each individual publication two individual nurse- and physiotherapist-sessions
indicated by at least 1 member of the Committee to have improved patient outcomes and reduced costs in a 12-
an impact on COPD management, and to reach a month follow-up.
consensus on the changes in the report. Disagreements Reference: Gallefoss F. The effects of patient education
were decided by vote. in COPD in a 1-year follow-up randomised, controlled
trial. Patient Educ Couns. 2004 Mar;52(3):259-66.
*Members: K. Rabe, Chair; P. Barnes, S. Buist, P. Calverley,
L. Fabbri, Y. Fukuchi, W. MacNee, R. Rodriguez-Roisin, I. Zielinski

vi
GOLD_WR_05 8/18/05 12:56 PM Page vii

2. Page 70 - Add sentence: Twenty-one days of inhaled An Appendix includes a report on outcome measures
tiotropium, 18 mg/day as a dry powder does not reduce for COPD to encourage comments and input from the
mucus clearance from the lungs. scientific community to prepare for the full revision of the
Reference: Hasani A, Toms N, Agnew JE, Sarno M, report, scheduled to appear in mid-2006. The many
Harrison AJ, Dilworth P. The effect of inhaled tiotropium individuals who participated in preparation of this report
bromide on lung mucociliary clearance in patients with are listed in the document. The GOLD Science
COPD. Chest. 2004 May;125(5):1726-34. Committee is grateful to those who contributed to this
report, particularly the work of Dr. Paul Jones, London,
3. Page 71 – Add sentence: Short-term treatment with England and Dr. Alvar Agusti, Palma de Mallorca, Spain.
a combined inhaled glucocorticosteroid and long-acting
ß2-agonist resulted in greater control of lung function and The proposed modifications for the GOLD Workshop
symptoms than combined anticholinergic and short-acting Report (Updated 2005) were approved by the GOLD
b2-agonist. Executive Committee.
Reference: Donohue JF, Kalberg C, Emmett A, Merchant
K, Knobil K. A short-term comparison of fluticasone The GOLD Workshop Report (Updated 2005), the GOLD
propionate/ salmeterol with ipratropium bromide/albuterol Executive Summary(Updated 2005), and the GOLD
for the treatment of COPD. Treat Respir Med. Pocket Guide (Updated 2005) along with the complete list
2004;3(3):173-81. of references examined by the Committee are available
on the GOLD website (www.goldcopd.org).
4. Page 73 - Change paragraph on immunoregulators to
read: Studies using an immunostimulator in COPD show
a decrease in the severity and frequency of exacerbations94
(add new reference). However, additional studies to
examine the long term effects of this therapy are required
before regular use can be recommended (Evidence B).
Reference: Li J, Zheng JP, Yuan JP, Zeng GQ, Zhong
NS, Lin CY. Protective effect of a bacterial extract against
acute exacerbation in patients with chronic bronchitis
accompanied by chronic obstructive pulmonary disease.
Chin Med J (Engl). 2004 Jun;117(6):828-34.

5. Page 93 - Add sentence: Early outpatient pulmonary


rehabilitation after hospitalization for COPD exacerbation
results in exercise capacity and health status improvements
at three months.
Reference: Man WD, Polkey MI, Donaldson N, Gray BJ,
Moxham J. Community pulmonary rehabilitation after
hospitalisation for acute exacerbations of chronic obstructive
pulmonary disease: randomised controlled study. BMJ.
2004 Nov 20;329(7476):1209.

A major new segment appears in Chapter 5-4 on antibiotics


in treatment of COPD exacerbations (page 94). The
material was prepared by the GOLD Science Committee
which gratefully acknowledges the opportunity to review
a statement on this topic prepared by the European
Respiratory Society and provided to the Committee by
Dr. William MacNee and Dr. Mark Woodhead. Prior to its
release, the material was reviewed by Dr. Sanjay Sethi,
State University of New York at Buffalo, Buffalo, New York,
and Dr. Antonio Anzueto, University of San Antonio, San
Antonio, Texas.

vii
GOLD_WR_05 8/18/05 12:56 PM Page viii

TABLE OF CONTENTS

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1 Inflammation and COPD Risk Factors . . . . . . .32


Proteinase-Antiproteinase Imbalance . . . . . . .32
1. Definition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5 Oxidative Stress . . . . . . . . . . . . . . . . . . . .33
Key Points . . . . . . . . . . . . . . . . . . . . . . . . . . .6 Pathology . . . . . . . . . . . . . . . . . . . . . . . . . .33
Definition . . . . . . . . . . . . . . . . . . . . . . . . . . .6 Central Airways . . . . . . . . . . . . . . . . . . . . .33
Natural History . . . . . . . . . . . . . . . . . . . . . . .7 Peripheral Airways . . . . . . . . . . . . . . . . . .34
Classification of Severity . . . . . . . . . . . . . . .7 Lung Parenchyma . . . . . . . . . . . . . . . . . . .35
Variable Course of COPD . . . . . . . . . . . . . .8 Pulmonary Vasculature . . . . . . . . . . . . . . .36
Scope of the Report . . . . . . . . . . . . . . . . . . .9 Pathophysiology . . . . . . . . . . . . . . . . . . . . .36
Asthma and COPD . . . . . . . . . . . . . . . . . . .9 Mucus Hypersecretion and
Pulmonary Tuberculosis and COPD . . . . . .9 Ciliary Dysfunction . . . . . . . . . . . . . . . . .36
References . . . . . . . . . . . . . . . . . . . . . . . . . .9 Airflow Limitation and
Pulmonary Hyperinflation . . . . . . . . . . . .36
2. Burden of COPD . . . . . . . . . . . . . . . . . . . . . . . . .11 Gas Exchange Abnormalities . . . . . . . . . . . .37
Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .12
Pulmonary Hypertension and
Introduction . . . . . . . . . . . . . . . . . . . . . . . . .12
Cor Pulmonale . . . . . . . . . . . . . . . . . . . .38
Epidemiology . . . . . . . . . . . . . . . . . . . . . . .12
Systemic Effects . . . . . . . . . . . . . . . . . . . .38
Prevalence . . . . . . . . . . . . . . . . . . . . . . . .12
Pathophysiology and the
Morbidity . . . . . . . . . . . . . . . . . . . . . . . . . .14
Mortality . . . . . . . . . . . . . . . . . . . . . . . . . .14 Symptoms of COPD . . . . . . . . . . . . . . . .38
Economic and Social Burden of COPD . . . .15 Pathology and Pathophysiology of
Economic Burden . . . . . . . . . . . . . . . . . . .15 Exacerbations . . . . . . . . . . . . . . . . . . . . . .39
Social Burden . . . . . . . . . . . . . . . . . . . . . .16 Pathology . . . . . . . . . . . . . . . . . . . . . . . . .39
References . . . . . . . . . . . . . . . . . . . . . . . . .17 Pathophysiology . . . . . . . . . . . . . . . . . . . .39
References . . . . . . . . . . . . . . . . . . . . . . . . .39
3. Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . .19
Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .20 5. Management of COPD . . . . . . . . . . . . . . . . . . . . .45
Introduction . . . . . . . . . . . . . . . . . . . . . . . . .20 Introduction . . . . . . . . . . . . . . . . . . . . . . . . .46
Host Factors . . . . . . . . . . . . . . . . . . . . . . . .21
Genes . . . . . . . . . . . . . . . . . . . . . . . . . . . .21 Component 1: Assess and Monitor Disease . . .47
Airway Hyperresponsiveness . . . . . . . . . . .21 Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .47
Lung Growth . . . . . . . . . . . . . . . . . . . . . . .21 Initial Diagnosis . . . . . . . . . . . . . . . . . . . . . .47
Exposures . . . . . . . . . . . . . . . . . . . . . . . . . .21 Assessment of Symptoms . . . . . . . . . . . . . . .47
Tobacco Smoke . . . . . . . . . . . . . . . . . . . .21 Medical History . . . . . . . . . . . . . . . . . . . . .49
Occupational Dusts and Chemicals . . . . . .22 Physical Examination . . . . . . . . . . . . . . . .50
Outdoor and Indoor Air Pollution . . . . . . . .22
Measurement of Airflow Limitation . . . . . . . .50
Infections . . . . . . . . . . . . . . . . . . . . . . . . .22
Assessment of Severity . . . . . . . . . . . . . . .51
Socioeconomic Status . . . . . . . . . . . . . . . .23
Additional Investigations . . . . . . . . . . . . . .52
References . . . . . . . . . . . . . . . . . . . . . . . . .23
Differential Diagnosis . . . . . . . . . . . . . . . .53
4. Pathogenesis, Pathology, and Ongoing Monitoring and
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . .27 Assessment . . . . . . . . . . . . . . . . . . . . . . .53
Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .28 Monitor Disease Progression and
Introduction . . . . . . . . . . . . . . . . . . . . . . . . .28 Development of Complications . . . . . . . .54
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . .28 Monitor Pharmacotherapy and
Inflammatory Cells . . . . . . . . . . . . . . . . . .29 Other Medical Treatment . . . . . . . . . . . . .55
Inflammatory Mediators . . . . . . . . . . . . . . .30 Monitor Exacerbation History . . . . . . . . . .55
Differences Between Inflammation in Monitor Comorbidities . . . . . . . . . . . . . . . .55
COPD and Asthma . . . . . . . . . . . . . . . . .31 References . . . . . . . . . . . . . . . . . . . . . . . . .56

viii
GOLD_WR_05 8/18/05 12:56 PM Page ix

Component 2: Reduce Risk Factors . . . . . . . . . .58 Non-Pharmacologic Treatment . . . . . . . . . . .74


Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .58 Rehabilitation . . . . . . . . . . . . . . . . . . . . . .74
Introduction . . . . . . . . . . . . . . . . . . . . . . . . .58 Oxygen Therapy . . . . . . . . . . . . . . . . . . . .76
Tobacco Smoke . . . . . . . . . . . . . . . . . . . . .58 Ventilatory Support . . . . . . . . . . . . . . . . . .77
Smoking Prevention . . . . . . . . . . . . . . . . .58 Surgical Treatments . . . . . . . . . . . . . . . . . .78
Smoking Cessation . . . . . . . . . . . . . . . . . .58 Special Considerations . . . . . . . . . . . . . . .79
Occupational Exposures . . . . . . . . . . . . . . .62 References . . . . . . . . . . . . . . . . . . . . . . . . .80
Indoor/Outdoor Air Pollution . . . . . . . . . . . .62
Regulation of Air Quality . . . . . . . . . . . . . .62 Component 4: Manage Exacerbations . . . . . .88
Patient-Oriented Control . . . . . . . . . . . . . .62 Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .88
References . . . . . . . . . . . . . . . . . . . . . . . . .63 Introduction . . . . . . . . . . . . . . . . . . . . . . . . .88
Diagnosis and Assessment of Severity . . . .88
Component 3: Manage Stable COPD . . . . . . . . .65 Medical History . . . . . . . . . . . . . . . . . . . . .88
Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .65 Assessment of Severity . . . . . . . . . . . . . . .89
Introduction . . . . . . . . . . . . . . . . . . . . . . . . .65 Home Management . . . . . . . . . . . . . . . . . . .89
Education . . . . . . . . . . . . . . . . . . . . . . . . . .65 Bronchodilator Therapy . . . . . . . . . . . . . . .90
Goals and Educational Strategies . . . . . . .66 Glucocorticosteroids . . . . . . . . . . . . . . . . .90
Components of an Education Program . . .66 Hospital Management . . . . . . . . . . . . . . . . .91
Cost Effectiveness of Education Emergency Department or Hospital . . . . . .91
Programs for COPD Patients . . . . . . . . . .67 Hospital Discharge and Follow-Up . . . . . . .93
Pharmacologic Treatment . . . . . . . . . . . . . .67 Antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . .94
Overview of the Medications . . . . . . . . . . .67 References . . . . . . . . . . . . . . . . . . . . . . . . .96
Bronchodilators . . . . . . . . . . . . . . . . . . . . .67
Glucocorticosteroids . . . . . . . . . . . . . . . . .71 6. Future Research . . . . . . . . . . . . . . . . . . . . . . . . . .99
Pharmacologic Therapy by Disease Severity . .72
Other Pharmacologic Treatments . . . . . . .73 APPENDIX
Outcomes and Markers in COPD . . . . . . . .103

ix
GOLD_WR_05 8/18/05 12:56 PM Page x

x
GOLD_WR_05 8/18/05 12:56 PM Page 1

INTRODUCTION
hronic Obstructive Pulmonary Disease (COPD) is the molecular and cellular mechanisms involved in COPD
C a major cause of chronic morbidity and mortality
throughout the world. Many people suffer from this
pathogenesis should lead to effective treatments that
slow or halt the course of the disease.
disease for years and die prematurely from it or its
complications. COPD is currently the fourth leading GOLD WORKSHOP REPORT:
cause of death in the world1, and further increases in its GLOBAL STRATEGY FOR THE
prevalence and mortality can be predicted in the coming
decades2. A unified international effort is needed to
DIAGNOSIS, MANAGEMENT, AND
reverse these trends. PREVENTION OF COPD

The Global Initiative for Chronic Obstructive Lung One strategy to help achieve GOLD's objectives is to
Disease (GOLD) is conducted in collaboration with the provide health care workers, health care authorities,
US National Heart, Lung, and Blood Institute (NHLBI) and the general public with state-of-the-art information
and the World Health Organization (WHO). Its goals are about COPD and specific recommendations on the most
to increase awareness of COPD and decrease morbidity appropriate management and prevention strategies.
and mortality from the disease. GOLD aims to improve The GOLD Workshop Report, Global Strategy for the
prevention and management of COPD through a concerted Diagnosis, Management, and Prevention of COPD, is
worldwide effort of people involved in all facets of health based on the best-validated current concepts of COPD
care and health care policy, and to encourage a renewed pathogenesis and the available evidence on the most
research interest in this highly prevalent disease. appropriate management and prevention strategies.
The Report has been developed by individuals with
A nihilistic attitude toward COPD has arisen among expertise in COPD research and patient care and exten-
some health care providers, due to the relatively limited sively reviewed by many experts and scientific societies.
success of primary and secondary prevention (i.e., It provides state-of-the-art information about COPD for
avoidance of factors that cause COPD or its progression), pulmonary specialists and other interested physicians.
the prevailing notion that COPD is largely a self-inflicted The document will also serve as a source for the produc-
disease, and disappointment with available treatment tion of various communications during the implementation
options. The GOLD project will work toward combating of the GOLD program, including a practical guide for
this nihilistic attitude by disseminating information about primary care physicians and a document for use in
available treatments, both pharmacologic and non- developing countries.
pharmacologic.
The GOLD Report is not intended to be a comprehensive
Tobacco smoking is a major cause of COPD, as well as textbook on COPD, but rather to summarize the current
of many other diseases. A decline in tobacco smoking state of the field. Each chapter starts with Key Points
would result in substantial health benefits and a decrease that crystallize current knowledge. The chapters on the
in the prevalence of COPD and other smoking-related Burden of COPD and Risk Factors demonstrate the
diseases. There is an urgent need for improved strategies global importance of COPD and the various causal
to decrease tobacco consumption. However, tobacco factors involved. The chapter on Pathogenesis,
smoking is not the only cause of COPD and may not Pathology, and Pathophysiology documents the current
even be the major cause in some parts of the world. understanding of, and remaining questions about, the
Furthermore, not all smokers develop clinically significant mechanism(s) that lead to COPD, as well as the
COPD, which suggests that additional factors are structural and functional abnormalities of the lungs
involved in determining each individual's susceptibility. characteristic of the disease.
Thus, investigation of COPD risk factors and ways to
reduce exposure to these factors is also an important A major part of the GOLD Workshop Report is devoted
area for future research. New research tools have to the clinical Management of COPD and presents a
recently revealed that inflammation plays a prominent management plan with four components: (1) Assess
role in COPD pathogenesis, but this inflammation is and Monitor Disease; (2) Reduce Risk Factors;
different than that involved in asthma. Further study of (3) Manage Stable COPD; (4) Manage Exacerbations.

INTRODUCTION 1
GOLD_WR_05 8/18/05 12:56 PM Page 2

Management recommendations are largely symptom In April 1998, the NHLBI and WHO cosponsored a work-
driven and are presented according to the severity of shop to begin the development of the Report. Workshop
the disease, using a simple classification of severity to participants were divided into three groups: definition and
facilitate the practical implementation of the available natural history, chaired by Dr. Sonia Buist; pathophysiology,
management options. Where appropriate, information risk factors, diagnosis, and classification of severity,
about health education for patients is included. chaired by Dr. Leonardo Fabbri; and management,
chaired by Dr. Romain Pauwels. A table of contents
The final chapter identifies critical gaps in knowledge was developed and writing assignments were made.
requiring Further Research and provides a summary of The Panel agreed that clinical recommendations would
proposed directions for the development of new therapeutic require scientific evidence, or would be clearly labeled as
approaches. "expert opinion." Each chapter would contain a set of the
most current and representative references.
METHODS USED TO DEVELOP
THIS REPORT In September 1998, the Panel met to evaluate its
progress. Members reviewed a variety of evidence tables
and chose to assign levels of evidence to statements
In January 1997, COPD experts from several countries using the system developed by the NHLBI (Figure A).
met in Brussels, Belgium to explore the development of a Levels of evidence are assigned to management
Global Initiative for Chronic Obstructive Lung Disease. recommendations where appropriate in Chapter 5,
Dr. Romain Pauwels served as Chair; representatives of Management of COPD, and are indicated in boldface
the NHLBI and WHO attended. Participants agreed that type enclosed in parentheses after the relevant statement
the project was timely and important, and recommended - e.g., (Evidence A). The methodological issues
the establishment of a panel with expertise on a wide concerning the use of evidence from meta-analyses were
variety of COPD-related topics to prepare an evidence- carefully considered (e.g., a meta-analysis of a number
based document on diagnosis, management, and of smaller studies considered to be evidence level B)2.
prevention of COPD. NHLBI and WHO staff, in concert The panel met in May 1999, September 1999, and May
with Dr. Pauwels, identified individuals from many regions 2000 in conjunction with meetings of the American
of the world to serve on the Expert Panel, which included Thoracic Society (ATS) and the European Respiratory
health professionals in the areas of respiratory medicine, Society (ERS). Symposia were held at these meetings to
epidemiology, pathology, socioeconomics, public health, present the developing program and to solicit opinion and
and health education. comments. The meeting in May 2000 was the final
consensus workshop.
The first step toward developing the Workshop Report
was to review the multiple COPD guidelines already After this workshop, the document was submitted for
published. The NHLBI collected these guidelines and review to individuals and medical societies interested in
prepared a summary table of similarities and differences the management of COPD. The reviewers' comments
between the documents. Where agreement existed, the were incorporated, as appropriate, into the final docu-
Expert Panel drew on these existing documents for use ment by the Chair in cooperation with members of the
in the Workshop Report. Where major differences existed, Expert Panel. Prior to its release for publication, the
the Expert Panel agreed to carefully examine the scientific Report was reviewed by the NHLBI and the WHO.
evidence to reach an independent conclusion. A workshop was held in September, 2000 to begin
implementation of the GOLD program.
In September 1997, several members of the Expert Panel
met with a consultant to develop a comprehensive set
of terms to build a database of COPD literature. The
database and a computer program to search the world
literature on COPD have been developed, and they will
be placed on the Internet and cross-referenced with the
Workshop Report to help keep the Report current as new
literature is published.

2 INTRODUCTION
GOLD_WR_05 8/18/05 12:56 PM Page 3

Figure A. Description of Levels of Evidence


Evidence Category Sources of Evidence Definition

A Randomized controlled Evidence is from endpoints of well-designed RCTs that


trials (RCTs). provide a consistent pattern of findings in the population
Rich body of data. for which the recommendation is made. Category A requires
substantial numbers of studies involving substantial numbers
of participants.

B Randomized controlled Evidence is from endpoints of intervention studies that


trials (RCTs). Limited include only a limited number of patients, posthoc or
body of data. subgroup analysis of RCTs, or meta-analysis of RCTs.
In general, Category B pertains when few randomized trials
exist, they are small in size, they were undertaken in a
population that differs from the target population of the
recommendation, or the results are somewhat inconsistent.

C Nonrandomized trials. Evidence is from outcomes of uncontrolled or nonrandomized


Observational studies. trials or from observational studies.

D Panel Consensus This category is used only in cases where the provision of
Judgment. some guidance was deemed valuable but the clinical literature
addressing the subject was deemed insufficient to justify
placement in one of the other categories. The Panel
Consensus is based on clinical experience or knowledge that
does not meet the above-listed criteria.

REFERENCES

1. World Health Organization. World health report. Geneva: World Health Organization; 2000.
Available from: URL: http://www.who.int/whr/2000/en/statistics.htm
2. Murray CJL, Lopez AD. Evidence-based health policy - lessons from the Global Burden of Disease Study.
Science 1996; 274:740-3.

INTRODUCTION 3
GOLD_WR_05 8/18/05 12:56 PM Page 4

4 INTRODUCTION
GOLD_WR_05 8/18/05 12:56 PM Page 5

CHAPTER

DEFINITION
GOLD_WR_05 8/18/05 12:56 PM Page 6

CHAPTER 1: DEFINITION
obstructive pulmonary disease (COPD), and asthma. The
definitions of these terms variably emphasize structure
KEY POINTS: and function and are often based on whether the term is
• COPD is a disease state characterized by airflow used for clinical or research purposes. For example,
limitation that is not fully reversible. The airflow epidemiologists have created terminology and criteria,
limitation is usually both progressive and associ- based on functional status, that can be monitored in
ated with an abnormal inflammatory response of population-based studies or studies of physicians'
the lungs to noxious particles or gases. diagnoses1,2.

Based on current knowledge, a working definition of COPD


• The four-stage classification of COPD severity is a disease state characterized by airflow limitation that
used throughout this report provides an is not fully reversible. The airflow limitation is usually both
educational tool and a general indication of the progressive and associated with an abnormal inflammatory
approach to management. This conceptual response of the lungs to noxious particles or gases.
framework also emphasizes that COPD is usually Symptoms, functional abnormalities, and complications of
progressive if exposure to the noxious agent is COPD can all be explained on the basis of this underlying
continued. inflammation and the resulting pathology (Figure 1-1).
• The characteristic symptoms of COPD are
cough, sputum production, and dyspnea upon Figure 1-1. Mechanisms Underlying Airflow
exertion. Limitation in COPD
• Chronic cough and sputum production often INFLAMMATION
precede the development of airflow limitation
by many years and these symptoms identify
individuals at risk of developing COPD.
Small airway disease Parenchymal destruction
• The focus of this Workshop Report is primarily
on COPD caused by inhaled particles and gases,
the most common of which worldwide is tobacco
smoke. AIRFLOW LIMITATION
• COPD can coexist with asthma, the other major
chronic obstructive airway disease characterized The chronic airflow limitation characteristic of COPD is
by an underlying airway inflammation. However, caused by a mixture of small airway disease (obstructive
the inflammation characteristic of COPD is distinct bronchiolitis) and parenchymal destruction (emphysema),
from that of asthma. the relative contributions of which vary from person to
person. Chronic inflammation causes remodeling and
• Pulmonary tuberculosis may affect lung function narrowing of the small airways. Destruction of the lung
and symptomatology and, in areas where parenchyma, also by inflammatory processes, leads to
tuberculosis is prevalent, can lead to confusion the loss of alveolar attachments to the small airways and
in the diagnosis of COPD. decreases lung elastic recoil; in turn, these changes diminish
the ability of the airways to remain open during expiration.
Airflow limitation is measured by spirometry, as this is the
DEFINITION most widely available, reproducible test of lung function.

For years, clinicians, physiologists, pathologists, and Many previous definitions of COPD have emphasized the
epidemiologists have struggled with the definitions of terms "emphysema" and "chronic bronchitis," which are
disorders associated with chronic airflow limitation, no longer included in the definition of COPD used in this
including chronic bronchitis, emphysema, chronic report. Emphysema, or destruction of the gas-exchanging

6 DEFINITION
GOLD_WR_05 8/18/05 12:56 PM Page 7

surfaces of the lung (alveoli), is a pathological term that some improvement in function and will certainly slow or
is often (but incorrectly) used clinically and describes even halt the progression of the disease.
only one of several structural abnormalities present in
patients with COPD. Chronic bronchitis, or the presence Classification of Severity: Stages of COPD
of cough and sputum production for at least 3 months in
each of two consecutive years, remains a clinically and For educational reasons, a simple classification of disease
epidemiologically useful term. However, it does not severity into four stages is recommended (Figure 1-2).
reflect the major impact of airflow limitation on morbidity The staging is based on airflow limitation as measured by
and mortality in COPD patients. It is also important to spirometry, which is essential for diagnosis and provides
recognize that cough and sputum production may precede a useful description of the severity of pathological
the development of airflow limitation; conversely, some changes in COPD. Specific FEV1 cut-points (e.g.,
patients develop significant airflow limitation without < 80% predicted) are used for purposes of simplicity:
chronic cough and sputum production. these cut-points have not been clinically validated.

NATURAL HISTORY The impact of COPD on an individual patient depends


not just on the degree of airflow limitation, but also on
COPD has a variable natural history and not all individuals the severity of symptoms (especially breathlessness and
follow the same course. However, COPD is generally a decreased exercise capacity) and complications of the
progressive disease, especially if a patient's exposure disease. The management of COPD is largely symptom
to noxious agents continues. If exposure is stopped, the driven, and there is only an imperfect relationship between
disease may still progress due to the decline in lung the degree of airflow limitation and the presence of
function that normally occurs with aging. Nevertheless, symptoms. The staging, therefore, is a pragmatic
stopping exposure to noxious agents, even after approach aimed at practical implementation and should
significant airflow limitation is present, can result in only be regarded as an educational tool, and a very

Figure 1-2. Classification of Severity of COPD

Stage Characteristics

0: At Risk • normal spirometry


• chronic symptoms (cough, sputum production)
I: Mild COPD • FEV1/FVC < 70%
• FEV1 ≥ 80% predicted
• with or without chronic symptoms (cough, sputum production)
II: Moderate COPD • FEV1/FVC < 70%
• 50% ≤ FEV1 < 80% predicted
• with or without chronic symptoms (cough, sputum production)
III: Severe COPD • FEV1/FVC < 70%
• 30% ≤ FEV1 < 50% predicted
• with or without chronic symptoms (cough, sputum production)
IV: Very Severe COPD • FEV1/FVC < 70%
• FEV1 < 30% predicted or FEV1 < 50% predicted plus chronic respiratory
failure

Classification based on postbronchodilator FEV1


FEV1: forced expiratory volume in one second; FVC: forced vital capacity; respiratory failure: arterial partial pressure of oxygen (PaO2) less than
8.0 kPa (60 mm Hg) with or without arterial partial pressure of CO2 (PaCO2) greater than 6.7 kPa (50 mm Hg) while breathing air at sea level.

DEFINITION 7
GOLD_WR_05 8/18/05 12:56 PM Page 8

general indication of the approach to management. All may also lead to effects on the heart such as cor
FEV1 values refer to postbronchodilator FEV1. pulmonale (right heart failure). Clinical signs of cor
pulmonale include elevation of the jugular venous pressure
Although COPD is defined on the basis of airflow and pitting ankle edema. Patients may have Stage IV:
limitation, in practice the decision to seek medical help Very Severe COPD even if the FEV1 is > 30% predicted,
(and so permit the diagnosis to be made) is normally whenever these complications are present. At this stage,
determined by the impact of a particular symptom on a quality of life is very appreciably impaired and exacerba-
patient's lifestyle. Thus, COPD may be diagnosed at any tions may be life threatening.
stage of the illness.
Variable Course of COPD
The characteristic symptoms of COPD are cough,
The common statement that only 15-20% of smokers
sputum production, and dyspnea upon exertion. Chronic
develop clinically significant COPD is misleading. A
cough and sputum production often precede the
much higher proportion develop abnormal lung function
development of airflow limitation by many years, although at some point if they continue to smoke. Not all individuals
not all individuals with cough and sputum production go with COPD follow the classical linear course as outlined
on to develop COPD. This pattern offers a unique in the Fletcher and Peto diagram, which is actually the
opportunity to identify those at risk for COPD and mean of many individual courses3.
intervene when the disease is not yet a health problem.
A major objective of GOLD is to increase awareness Figure 1-3 shows four examples of the various courses
among health care providers and the general public of that individual COPD patients may follow. Panel A
the significance of these symptoms. illustrates an individual who has cough and sputum
production, but never develops abnormal lung function
Stage 0: At Risk - Characterized by chronic cough and (as defined in this Report). Panel B illustrates an
sputum production. Lung function, as measured by individual who develops abnormal lung function but who
spirometry, is still normal. may never come to diagnosis. Panel C illustrates a
person who develops abnormal lung function around
Stage I: Mild COPD - Characterized by mild airflow age 50, then progressively deteriorates over about 15
limitation (FEV1/FVC < 70% but FEV1 > 80% predicted) years and dies of respiratory failure at age 65. Panel D
and usually, but not always, by chronic cough and sputum illustrates an individual who develops abnormal lung
production. At this stage, the individual may not even be function in mid-adult life and continues to deteriorate
aware that his or her lung function is abnormal. This gradually but never develops respiratory failure and does
underscores the importance of health care providers not die as a result of COPD.
doing spirometry in all smokers so that their lung function
can be observed and recorded over time.

Stage II: Moderate COPD - Characterized by worsening


Figure 1-3. Examples of Individual Patient Histories
airflow limitation (50% < FEV1 < 80% predicted), and
usually the progression of symptoms with shortness of
breath typically developing on exertion. This is the stage Age Age
at which patients typically seek medical attention
because of dyspnea or an exacerbation of their disease.

Stage III: Severe COPD - Characterized by further


worsening of airflow limitation (30% ≤ FEV1 < 50% pre-
A B
dicted), increased shortness of breath, and repeated
exacerbations which have an impact on patients’ quality
of life. Age Age

Stage IV: Very Severe COPD - Characterized by severe


airflow limitation (FEV1 < 30% predicted) or the presence
of chronic respiratory failure. Respiratory failure is
defined as an arterial partial pressure of O2 (PaO2) less
than 8.0 kPa (60 mmHg) with or without arterial partial C D
pressure of CO2 (PaCO2) greater than 6.7 kPa (50 mm
Hg) while breathing air at sea level. Respiratory failure

8 DEFINITION
GOLD_WR_05 8/18/05 12:56 PM Page 9

better understood it will remain difficult to differentiate the


SCOPE OF THE REPORT two diseases in some individual patients. Given the current
state of medical and scientific knowledge, an attempt to
The focus of this Report is primarily on COPD caused by determine an absolutely rigid definition of COPD or asthma
inhaled particles and gases, the most common of which is bound to end up in semantics.
worldwide is tobacco smoke. Poorly reversible airflow
limitation associated with bronchiectasis, cystic fibrosis, Pulmonary Tuberculosis and COPD
tuberculosis, or asthma is not included except insofar as
these conditions overlap with COPD. In many developing countries both pulmonary tuberculosis
and COPD are common. In countries where tuberculosis
Asthma and COPD is very common, respiratory abnormalities may be too
readily attributed to this disease. Conversely, where the
COPD can coexist with asthma, the other major chronic rate of tuberculosis is greatly diminished, the possible
obstructive airway disease characterized by an underlying diagnosis of this disease is sometimes overlooked.
airway inflammation. Asthma and COPD have their major
symptoms in common, but these are generally more Chronic bronchitis/bronchiolitis and emphysema often
variable in asthma than in COPD. The underlying chronic occur as complications of pulmonary tuberculosis and
airway inflammation is also very different (Figure 1-4): are important contributors to the mixed lung function
that in asthma is mainly eosinophilic and driven by CD4+ changes characteristic of tuberculosis4. The degree of
T lymphocytes, while that in COPD is neutrophilic and obstructive airway changes5 in treated patients with
characterized by the presence of increased numbers of pulmonary tuberculosis increases with age, the amount
macrophages and CD8+ T lymphocytes. In addition, of cigarettes smoked, and the extent of the initial
airflow limitation in asthma is often completely reversible, tuberculous disease6. In patients with both diseases,
either spontaneously or with treatment, while in COPD it is COPD adds to the disability of pulmonary tuberculosis,
never fully reversible and is usually progressive if exposure and vice versa.
to noxious agents continues. Finally, the responses to
treatment of asthma and COPD are dramatically different, Therefore, in all subjects with symptoms of COPD, a
in terms of both the overall magnitude of the achievable possible diagnosis of tuberculosis should be considered,
response and the qualitative effects of specific treatments especially in areas where this disease is known to be
such as anticholinergics and glucocorticosteroids. prevalent. Investigations to exclude tuberculosis should
However, there is undoubtedly an overlap between be a routine part of COPD diagnosis, the intensity of the
asthma and COPD. Individuals with asthma who are diagnostic procedures depending on the degree of
exposed to noxious agents that cause COPD may develop suspicion. Chest radiograph and sputum culture are
a mixture of "asthma-like" inflammation and "COPD-like" helpful in making the differential diagnosis.
inflammation. There is also evidence that longstanding
asthma on its own can lead to airway remodeling and
partly irreversible airflow limitation. Asthma can usually REFERENCES
be distinguished from COPD, but until the causal mecha-
nisms and pathognomonic markers of these diseases are 1. Samet JM. Definitions and methodology in COPD
research. In: Hensley M, Saunders N, eds. Clinical epi-
Figure 1-4. Asthma and COPD demiology of chronic obstructive pulmonary disease. New
York: Marcel Dekker; 1989. p. 1-22.
Asthma COPD 2. Vermeire PA, Pride NB. A "splitting" look at chronic non-
Sensitizing agent Noxious agent specific lung disease (CNSLD): common features but
diverse pathogenesis. Eur Respir J 1991; 4:490-6.
3. Fletcher C, Peto R. The natural history of chronic airflow
Asthmatic airway inflammation COPD airway inflammation obstruction. BMJ 1977; 1:1645-8.
CD4+ T lymphocytes CD8+ T lymphocytes 4. Leitch AG. Pulmonary tuberculosis: clinical features. In:
Eosinophis Macrophages Neutrophils
Crofton J, Douglas A, eds. Respiratory diseases. Oxford:
Blackwell Science; 2000. p. 507-27.
5. Birath G, Caro J, Malmberg R, Simonsson BG. Airway
obstruction in pulmonary tuberculosis. Scand J Resp Dis
Completely Completely 1966; 47:27-36.
reversible Airflow Limitation irreversible
6. Snider GL, Doctor L, Demas TA, Shaw AR. Obstructive air-
way disease in patients with treated pulmonary tuberculo-
sis. Am Rev Respir Dis 1971; 103:625-40.

DEFINITION 9
GOLD_WR_05 8/18/05 12:56 PM Page 10

10 DEFINITION
GOLD_WR_05 8/18/05 12:56 PM Page 11

CHAPTER

BURDEN OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 12

CHAPTER 2: BURDEN OF COPD


KEY POINTS:
EPIDEMIOLOGY
• COPD prevalence and morbidity data that are Most of the information available on COPD prevalence,
available probably greatly underestimate the total morbidity, and mortality comes from developed countries.
burden of the disease because it is not usually Even in these countries, accurate epidemiological data
recognized and diagnosed until it is clinically on COPD are difficult and expensive to collect.
apparent and moderately advanced. Prevalence and morbidity data greatly underestimate the
total burden of COPD because the disease is usually not
• Prevalence, morbidity, and mortality vary diagnosed until it is clinically apparent and moderately
appreciably across countries, but in all countries advanced. The imprecise and variable definitions of
where data are available COPD is a significant COPD have made it hard to quantify the morbidity and
health problem in both men and women. mortality of this disease in developed1 and developing
countries. Mortality data also underestimate COPD as a
• The substantial increase in the global burden cause of death because the disease is more likely to be
of COPD projected over the next twenty years cited as a contributory than as an underlying cause of
reflects, in large part, the increasing use of death, or may not be cited at all2.
tobacco worldwide, and the changing age
structure of populations in developing countries. Prevalence
• Medical expenditures for treating COPD and Available estimates of COPD prevalence have been
the indirect costs of morbidity can represent a developed by determining either the proportion of the
substantial economic and social burden for population that reports having respiratory symptoms
societies and public and private payers and/or airflow limitation, or the proportion that reports
worldwide. Nevertheless, very little economic having been diagnosed with COPD, chronic bronchitis, or
information concerning COPD is available. emphysema by a physician. Each of these approaches
will yield a different estimate, and may be useful for
different purposes. For example, studies that ask about
INTRODUCTION the full range of COPD symptoms from early to advanced
disease are useful to estimate the total societal burden of
COPD is a leading cause of morbidity and mortality the disease. Data on doctor diagnoses of COPD are
worldwide and results in an economic and social burden useful to estimate the prevalence of clinically significant
that is both substantial and increasing. COPD prevalence, disease that is of sufficient severity to require health
morbidity, and mortality vary appreciably across countries services, and therefore is likely to incur significant costs.
and across different groups within countries, but in general
are directly related to the prevalence of tobacco smoking. The population surveys necessary to develop accurate
Most epidemiological studies have found that COPD estimates of COPD prevalence are costly to do and
prevalence, morbidity, and mortality have increased over therefore have not been conducted in many countries.
time and are greater in men than in women. Very few Obtaining reliable prevalence data for COPD in each
studies have quantified the economic and social burden country should be a priority in order to alert those
of COPD. In developed countries, the direct medical responsible for planning prevention services and health
costs of COPD are substantial because the disease is care delivery to the high prevalence and cost of the
both chronic and highly prevalent. In developing disease. The prevalence of COPD is likely to vary
countries, the indirect cost of COPD from loss of work appreciably depending on the prevalence of risk
and productivity may be more important than the direct factor exposure, age distribution, and prevalence of
costs of medical care. susceptibility genes in different countries.

Until recently, virtually all population-based studies


in developed countries showed a markedly greater
prevalence and mortality of COPD among men

12 BURDEN OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 13

compared to women3-6. Gender-related differences in In the NHANES 3 study16, airflow limitation was defined
exposure to risk factors, mostly cigarette smoking, as an FEV1/FVC < 70%. The prevalence of airflow
probably explain this pattern. In developing countries, limitation was lower than the prevalence of respiratory
some studies report a slightly higher prevalence of COPD symptoms found in the same study, but both sets of data
in women than men. This likely reflects exposure to reinforce the view that smoking is the most important
indoor air pollution from cooking and heating fuels determinant of COPD prevalence in developed countries.
(greater among women) as well as exposure to tobacco Among white males, airflow limitation was present in
smoke (greater among men)7-15. Recent large population- 14.2% of current smokers, 6.9% of ex-smokers, and 3.3%
based studies in the US show a different pattern emerging, of never smokers. Among white females, the prevalence
with the prevalence of COPD almost equal in men and of airflow limitation was 13.6% in smokers, 6.8% in ex-
women16,17. This likely reflects the changing pattern of smokers, and 3.1% in never smokers. Airflow limitation
exposure to the most important risk factor, tobacco was more common among white smokers than among
smoke. black smokers.

Estimates based on self-report of respiratory symptoms. Estimates based on physician diagnosis of COPD.
COPD prevalence data based on self-report of respiratory COPD prevalence data based on physician diagnosis
symptoms (chronic cough, sputum production, wheezing, provide information about the prevalence of clinically
and shortness of breath) include people at risk for COPD significant COPD that is of sufficient severity to prompt a
(Stage 0) as well as those with airflow limitation, and thus visit to a physician. Few population-based prevalence
yield maximum prevalence estimates. These studies surveys have been published to provide this information,
reveal sizable variations in the prevalence of respiratory and available data are often confusing because asthma
symptoms depending on smoking status, age, and COPD diagnoses are not separated, all age groups
occupational and environmental exposures, country or are considered together, or chronic bronchitis and
region, and, to a lesser extent, gender and race. The emphysema are considered separately.
data also reveal appreciable variations over time, reflecting
important temporal changes in populations' exposure to In the UK the General Practice Research Database18,
risk factors such as smoking, outdoor air pollution, and which is based on 525 practices serving 3.4 million
occupational exposures. patients (6.4% of the total population of England and
Wales), provides population-based data on physician-
The third National Health and Nutrition Examination diagnosed COPD (Figure 2-1). In 1997, the prevalence
Survey (NHANES 3)16, a large national survey conducted of COPD was 1.7% among men and 1.4% among
in the US between 1988 and 1994, included self-report women. Between 1990 and 1997, the prevalence
questions about respiratory symptoms. The prevalence increased by 25% in men and 69% in women. The
of respiratory symptoms varied markedly by smoking prevalence of COPD among men plateaued in the mid-
status (current>ex>never). Among white males, chronic 1990s, but continued to increase among women, reaching
cough was reported by 24% of smokers, 4.7% of ex-
smokers, and 4.0% of never smokers. The prevalence Figure 2-1. Prevalence (%) of Physician-Diagnosed
of chronic cough among white women was 20.6% in COPD in the UK From 1990 to 1997 by Sex18
smokers, 6.5% in ex-smokers, and 5.0% in never smokers.
There was a smaller gradient in the prevalence of chronic 2.0
cough by race (white>black). The prevalence of sputum
production was similar to that of chronic cough in these
1.5
groups.

Estimates based on the presence of airflow limitation. 1.0


People may have respiratory symptoms such as cough
and sputum production for many years before developing
0.5
airflow limitation. Thus, COPD prevalence data based on
the presence of airflow limitation provide a more accurate
estimate of the burden of COPD that is, or probably soon 0.0
1990 1991 1992 1993 1994 1995 1996 1997
will be, clinically significant. However, the use of different
cut points to define airflow limitation makes comparing Men Women
the results of different studies difficult. Reprinted with permission from Soriano JR, Maier WC, Egger R, Visick G, Thakrar B, Sykes J,
et al. Thorax 2000; 55:789-94. Copyright 2000 BMJ Publishing Group.

BURDEN OF COPD 13
GOLD_WR_05 8/18/05 12:56 PM Page 14

in 1997 the level observed in men in 1990. The General The Global Burden of Disease study reported a
Practice Research Database includes all ages and thus significantly higher prevalence of COPD in China than in
underestimates the true impact of COPD on older adults. most of the other regions (26.20/1,000 among men and
23.70/1,000 among women). A more recent survey
The Global Burden of Disease Study. The WHO/World conducted in three regions of China (Northern: Beijing;
Bank Global Burden of Disease Study19,20 used data from Northeast: Liao-Ning; and South-Mid: HuBei) in persons
both published and unpublished studies to estimate the older than 15 years estimated the prevalence of COPD at
prevalence of various diseases in different countries and 4.21/1,000 among men and 1.84/1,000 among women21.
regions around the world (Figure 2-2). Where few data
for a region were available, experts made informed Morbidity
estimates. Where no information was available, preliminary
estimates were derived from data from other regions that Morbidity includes physician visits, emergency
were believed to have similar epidemiological patterns. department visits, and hospitalizations. COPD data-
Using this approach, the worldwide prevalence of COPD bases for these outcome parameters are less readily
in 1990 was estimated at 9.34/1,000 in men and available and usually less reliable than mortality data-
7.33/1,000 in women. However, these estimates include bases. The limited data available indicate that morbidity
all ages and underestimate the true prevalence of COPD due to COPD increases with age and is greater in men
in older adults. than women17,22,23.

Figure 2-2. COPD Around the World (All Ages)19,20 In the UK, general practice consultations for COPD
during one year ranged from 4.17/1,000 in 45- to
Region or Country 1990 Prevalence per 1,000
Males/Females 64-year-olds to 8.86/1,000 in 65- to 74-year-olds to
10.32/1,000 in 75- to 84-year-olds. These rates are
Established Market Economies 6.98/3.79
2 to 4 times the equivalent rates for chest pain due to
Formerly Socialist Economies of Europe 7.35/3.45
ischemic heart disease24.
India 4.38/3.44
China* 26.20/23.70
In 1994, according to statistics from the UK Office of
Other Asia and Islands 2.89/1.79
National Statistics25, there were 203,193 hospital
Sub-Saharan Africa 4.41/2.49
admissions in Northern Ireland, Scotland, Wales, and
Latin America and Caribbean 3.36/2.72
Middle Eastern Crescent 2.69/2.83
England for COPD; the average length of hospital stay
World 9.34/7.33
among those admitted for a COPD diagnosis was
9.9 days.
*The prevalence of COPD in China reported in this study has been
questioned based on recent publications from China21 - see text.
US data indicate that in 1997 there were 16.365 million
Given the striking dearth of population-based data on (60.6/1,000) ambulatory care visits for COPD and
COPD prevalence in many countries of the world, the 448,000 (1.66/1,000) hospitalizations for which COPD
values listed in Figure 2-2 should not be viewed as very was the first-listed discharge diagnosis23. Hospitalization
precise. Nevertheless, some general patterns emerge. rates for COPD increased with age and were higher
The prevalence of COPD is highest in countries where among men than among women. These data should
cigarette smoking has been, or still is, very common, be interpreted cautiously, however, because the ICD-9
while the prevalence is lowest in countries where smoking codes for COPD that were in use in 1997, 490-492 and
is less common, or total tobacco consumption per capita 494-496, include "bronchitis not specified as acute or
is still low. The lowest COPD prevalence among men chronic." Therefore, the data for ambulatory care visits
(2.69/1,000) was found in the Middle Eastern Crescent are likely to have been inflated by inclusion of visits for
(a group of 36 countries in North Africa and the Middle acute bronchitis16.
East) and the lowest prevalence among women
(1.79/1,000) was found in the region referred to as "Other Mortality
Asia and Islands" (a group of 49 countries and islands,
the largest of which is Indonesia and which includes Of all of the descriptive epidemiological data for COPD,
Papua New Guinea, Nepal, Vietnam, Korea, Hong Kong, mortality data are the most readily available, and
and many small island countries). Except in the Middle probably the most reliable. (The World Health
Eastern Crescent, the prevalence of COPD is higher Organization publishes mortality statistics for selected
among men than among women. causes of death annually for all WHO regions26;

14 BURDEN OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 15

additional information is available from the WHO ECONOMIC AND SOCIAL


Evidence for Health Policy Department27.) However,
inconsistent use of terminology for COPD causes
BURDEN OF COPD
problems that do not arise for many other diseases.
For example, prior to about 1968 and the Eighth Because COPD is highly prevalent and can be severely
Revision of the ICD, the terms "chronic bronchitis" and disabling, direct medical expenditures and the indirect
"emphysema" were used extensively. During the costs of morbidity and premature mortality from COPD
1970s, the term "COPD" increasingly replaced those can represent a substantial economic and social burden
terms in the US and some but not all other countries, for societies and public and private insurance payers
making comparisons of COPD mortality in different worldwide. Nevertheless, very little quantitative informa-
countries very difficult. However, the situation has tion concerning the economic and social burden of COPD
improved with the Ninth and Tenth Revisions of the is available in the literature today.
ICD, in which deaths from COPD or chronic airways
obstruction are included in the broad category of Economic Burden
"COPD and allied conditions" (ICD-9 codes 490-496
and ICD-10 codes J42-46). Cost of illness studies provide insight into the economic
impact of a disease. Some countries attempt to separate
The age-adjusted death rates for COPD by race and economic burden into disease-attributable direct and indi-
sex in the US from 1960 to 1996 by ICD code are rect costs. The direct cost is the value of health care
shown in Figure 2-317. COPD death rates are very resources devoted to diagnosis and medical management
low among people under age 45 in the US, but then of the disease. Indirect costs reflect the monetary
increase with age, and COPD becomes the fourth or consequences of disability, missed work and school,
fifth leading cause of death among those over 4517, a premature mortality, and caregiver or family costs resulting
pattern that reflects the cumulative effect of cigarette from the illness. Data on these topics from developing
smoking28. Although appreciable variations in mortality countries are not available, but data from the US and
across developed countries for both genders have been some European countries provide an understanding of
reported29, these differences should be interpreted the economic burden of COPD in developed countries.
cautiously. Differences between countries in death
certification, diagnostic practices, the structure of United States. Figure 2-4 compares the estimated costs
health care systems, and life expectancy have an of various lung disorders in the US in 1993. In 1993, the
appreciable impact on reported mortality rates. annual economic burden of COPD in the US was estimated
at $23.9 billion17, including $14.7 billion in direct expenditures
for medical care services, $4.7 billion in indirect morbidity
costs, and $4.5 billion in indirect costs related to premature
mortality. With an estimated 15.7 million cases of COPD
Figure 2-3. Age-Adjusted* Death Rates for
Chronic Obstructive Pulmonary Disease by in the US30, the estimated direct cost of COPD is $1,522
Race and Sex, US 1960-199617 per COPD patient per year.

Rate/100,000 Population
ICD/7 ICD/8 ICD/9
Figure 2-4. Direct and Indirect Costs of
Lung Diseases, 1993 (US $ Billions)17
White Male
Condition Total Direct Mortality- Morbidity- Total
Black Male**
Cost Medical Related Related Indirect
Cost Indirect Indirect Cost
Cost Cost
White Female
COPD 23.9 14.7 4.5 4.7 9.2
Black Female** Asthma 12.6 9.8 0.9 0.9 2.8
Influenza 14.6 1.4 0.1 13.1 13.2
Pneumonia 7.8 1.7 4.6 1.5 6.1
Tuberculosis 1.1 0.7 - - 0.4
1960 62 64 66 68 70 72 74 76 78 80 82 84 86 88 90 92 94 1996
*Age-adjusted to the 2000 standard **Nonwhite from 1960 to 1967.
Lung Cancer 25.1 5.1 17.1 2.9 20.0

BURDEN OF COPD 15
GOLD_WR_05 8/18/05 12:56 PM Page 16

In a US study31 of COPD-related illness costs based on including: disease prevalence and demographics, particu-
the 1987 National Medical Expenditure Survey, per capita larly smoking patterns; the type and usage patterns of
expenditures for inpatient hospitalizations of COPD health care and non-health care services among patients
patients ($5,409 per hospitalization) were 2.7 times the with COPD; the relative prices of health care services;
expenditures for patients without COPD ($2,001 per employment and wage rates; and the availability of
hospitalization). In 1992, under Medicare, the US medical prevention strategies and treatments for COPD.
government health insurance program for individuals Similar data from developing countries are not available.
over 65, annual per capita expenditures for people with
COPD ($8,482) were nearly 2.5 times higher than annual Figure 2-5. Four-Country Comparison
expenditures for people without COPD ($3,511)32. of COPD Direct and Indirect Costs
Country (ref) Year Direct Cost Indirect Cost Total Per Capita*
United Kingdom. In 1996, the direct cost of COPD in (US$ Millions) (US$ Millions) (US$ Millions) (US$)

the UK was approximately £846 million (about US $1.393 UK33 1996 778 3,312 4,090 65
billion) or £1,154 (about US $1,900) per person per year, The Netherlands34 1993 256 N/A N/A N/A#
according to data from the National Health Service (NHS) Sweden35 1991 179 281 460 60
US1 1993 14,700 9,200 23,900 87
Executive33. Pharmaceutical expenditures for COPD and
allied conditions accounted for 11.0% of the total expen- * Per capita valuation based on 1993 population estimates from the United
Nations Population Council and expressed in 1993 US dollars.
ditures for prescription medications. Only 2% of total # The authors did not provide estimates of indirect costs.
primary care expenditures were for COPD-related visits.
Home care. Individuals with COPD frequently receive
In 1996, lost work productivity, disability, and premature professional medical care in their homes. In some
mortality from COPD in the UK accounted for an estimated countries, national health insurance plans provide coverage
24 million days of work lost. The indirect cost of the for oxygen therapy, visiting nursing services, rehabilitation,
disease was estimated at £600 million (about US $960 and even mechanical ventilation in the home, although
million) for attendance and disability living allowance and coverage for specific services varies from country to
£1.5 billion (about US $2.4 billion) to employers for work country36.
absence and reduced productivity 24.
Any estimate of direct medical expenditures for home
The Netherlands. In 1993, the direct cost of COPD in care underrepresents the true cost of home care to society,
the Netherlands was estimated to exceed US $256 million, because it ignores the economic value of the care provided
or US $813 per patient per year. Assuming constant costs to those with COPD by family members. In developing
and treatment patterns, the direct cost is expected to reach countries especially, direct medical costs may be less
US $410 million per year by 2010. In 1993 inpatient important than the impact of COPD on workplace and
hospitalizations accounted for 57% of the total direct cost home productivity. Because the health care sector
of COPD, and medications accounted for an additional might not provide long-term supportive care services for
23%. The indirect cost of COPD in the Netherlands was severely disabled individuals, COPD may force two
not available34. individuals to leave the workplace - the affected individual
and a family member who must now stay home to care
Sweden. The direct cost of COPD-related medical care for the disabled relative. Since human capital is often the
in Sweden was estimated at 1.085 billion SEK (about US most important national asset for developing countries,
$179.4 million) in 1991. The estimated indirect cost of COPD may represent a serious threat to their economies.
COPD was an additional 1.699 billion SEK (about US
$280.8 million)35. Social Burden

Comparison of different countries. Figure 2-5 Since mortality offers a limited perspective on the human
provides data on the economic burden of COPD in four burden of a disease, it is desirable to find other measures
countries with Western styles of medical practice and of disease burden that are consistent and measurable
social or private insurance structures. The data are across nations. The World Bank/WHO Global Burden of
standardized to equivalent year on a per capita basis. Disease Study19 designed a method to estimate the fraction
After adjusting to a common base year and population, of mortality and disability attributable to major diseases
the costs of COPD were relatively similar. The remaining and injuries using a composite measure of the burden of
variability in across-country estimates of economic each health problem, the Disability-Adjusted Life Year
burden can be partly explained by several factors, (DALY). The DALYs for a specific condition are the sum

16 BURDEN OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 17

Figure 2-6. Leading Causes of Disability-Adjusted Life Years 5. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking,
(DALYs) Lost Worldwide, 1990 and 2020 (Projected)19,20 changes in smoking habits, and rate of decline in FEV1:
Percent of Percent of new insight into gender differences. Eur Respir J 1994;
Disease Rank Rank
Total Total 7:1056-61.
or Injury 1990 2020
DALYs DALYs
6. Feinleib M, Rosenberg HM, Collins JG, Delozier JE,
Lower respiratory infections 1 8.2 6 3.1 Pokras R, Chevarley FM. Trends in COPD morbidity and
Diarrheal diseases 2 7.2 9 2.7 mortality in the United States. Am Rev Respir Dis 1989;
Perinatal period conditions 3 6.7 11 2.5 140:S9-18.
Unipolar major depression 4 3.7 2 5.7
7. Chen JC, Mannino MD. Worldwide epidemiology of chronic
Ischemic heart disease 5 3.4 1 5.9
Cerebrovascular disease 6 2.8 4 4.4 obstructive pulmonary disease. Current Opinion in
Tuberculosis 7 2.8 7 3.1 Pulmonary Medicine 1999; 5:93-9.
Measles 8 2.6 25 1.1 8. Dossing M, Khan J, al-Rabiah F. Risk factors for chronic
Road traffic accidents 9 2.5 3 5.1 obstructive lung disease in Saudi Arabia. Respiratory Med
Congenital anomalies 10 2.4 13 2.2 1994; 88:519-22.
Malaria 11 2.3 19 1.5
9. Dennis R, Maldonado D, Norman S, Baena E, Martinez G.
COPD 12 2.1 5 4.1
Trachea, bronchus, lung cancer 33 0.6 15 1.8 Woodsmoke exposure and risk for obstructive airways dis-
Excerpted with permission from Murray CJL, Lopez AD. Science 1999; 274:740-3. ease among women. Chest 1996; 109:115-9.
Copyright 1999 American Association for the Advancement of Science.
10. Perez-Padilla R, Regalado U, Vedal S, Pare P, Chapela R,
Sansores R, et al. Exposure to biomass smoke and chron-
of years lost because of premature mortality and years ic airway disease in Mexican women. Am J Respir Crit
of life lived with disability, adjusted for the severity of Care Med 1996; 154:701-6.
disability. 11. Behera D, Jindal SK. Respiratory symptoms in Indian women
using domestic cooking fuels. Chest 1991; 100:385-8.
The leading causes of DALYs lost worldwide in 1990 12. Amoli K. Bronchopulmonary disease in Iranian housewives
and 2020 (projected) are shown in Figure 2-6. In 1990, chronically exposed to indoor smoke. Eur Respir J 1998;
COPD was the twelfth leading cause of DALYs lost in the 11:659-63.
world, responsible for 2.1% of the total. According to the 13. Pandey MR. Prevalence of chronic bronchitis in a rural
projections, COPD will be the fifth leading cause of community of the Hill Region of Nepal. Thorax 1984;
DALYs lost worldwide in 2020, behind ischemic heart 39:331-6.
disease, major depression, traffic accidents, and cere- 14. Pandey MR. Domestic smoke pollution and chronic bron-
brovascular disease. This substantial increase in the chitis in a rural community of the Hill Region of Nepal.
global burden of COPD projected over the next twenty Thorax 1984; 39:337-9.
years reflects, in large part, the increasing use of tobacco 15. Samet JM, Marbury M, Spengler J. Health effects and
worldwide and the changing age structure of populations sources of indoor air pollution. Am Rev Respir Dis 1987;
in developing countries. 136:1486-508.
16. National Center for Health Statistics. Current estimates from
the National Health Interview Survey, United States, 1995.
REFERENCES Washington, DC: Department of Health and Human
Services, Public Health Service, Vital and Health Statistics;
1. Pride NB, Vermeire P, Allegra L. Diagnostic labels applied 1995. Publication No. 96-1527.
to model case histories of chronic airflow obstruction. 17. National Heart, Lung, and Blood Institute. Morbidity & mor-
Responses to a questionnaire in 11 North American and tality: chartbook on cardiovascular, lung, and blood diseases.
Western European countries. Eur Respir J 1989; 2:702-9. Bethesda, MD: US Department of Health and Human
2. Mannino DM, Brown C, Giovino GA. Obstructive lung dis- Services, Public Health Service, National Institutes of Health;
ease deaths in the United States from 1979 through 1993. 1998. Available from: URL:
An analysis using multiple-cause mortality data. Am J www.nhlbi.nih.gov/nhlbi/seiin/other/cht-book/htm
Respir Crit Care Med 1997; 156:814-8. 18. Soriano JR, Maier WC, Egger P, Visick G, Thakrar B, Sykes
3. Buist AS, Vollmer WM. Smoking and other risk factors. In: J, et al. Recent trends in physician diagnosed COPD in
Murray JF, Nadel JA, eds. Textbook of respiratory women and men in the UK. Thorax 2000; 55:789-94.
medicine. Philadelphia: WB Saunders; 1994. p. 1259-87. 19. Murray CJL, Lopez AD. Evidence-based health policy -
4. Thom TJ. International comparisons in COPD mortality. Am lessons from the Global Burden of Disease Study. Science
Rev Respir Dis 1989; 140:S27-34. 1996; 274:740-3.

BURDEN OF COPD 17
GOLD_WR_05 8/18/05 12:56 PM Page 18

20. Murray CJL, Lopez AD, eds. The global burden of disease: a 29. Incalzi RA, Fuso L, De Rosa M, Forastiere F, Rapiti E,
comprehensive assessment of mortality and disability from Nardecchia B, et al. Co-morbidity contributes to predict mor-
diseases, injuries and risk factors in 1990 and projected to tality of patients with chronic obstructive pulmonary disease.
2020. Cambridge, MA: Harvard University Press; 1996. Eur Respir J 1997; 10:2794-800.
21. Xian Sheng Chen. Analysis of basic data of the study on 30. Singh GK, Matthews TJ, Clarke SC. Annual summary of
prevention and treatment of COPD. Chin J Tuber Respiratory births, marriages, divorces, and deaths: United States, 1994.
Dis 1998; 21:749-52 (with English abstract). Monthly Vital Statistics Report 14 (13). National Center for
22. Higgins MW, Thom T. Incidence, prevalence, and mortality: Health Statistics, Hyattsville, MD.
intra- and inter-country differences. In: Hensley M, Saunders 31. Sullivan SD, Strassels S, Smith DH. Characterization of the
N, eds. Clinical epidemiology of chronic obstructive pul- incidence and cost of COPD in the US. Eur Respir J 1996;
monary disease. New York: Marcel Dekker; 1989. p. 23-43. 9:S421.
23. National Center for Health Statistics. National hospital inter- 32. Grasso ME, Weller WE, Shaffer TJ, Diette GB, Anderson GF.
view survey. Vital and health statistics, series 10 (issues from Capitation, managed care, and chronic obstructive pul-
1974 to 1995). monary disease. Am J Respir Crit Care Med 1998; 158:133-8.
24. Calverley PMA. Chronic obstructive pulmonary disease: the 33. National Health Service Executive. Burdens of disease: a dis-
key facts. London: British Lung Foundation; 1998. cussion document. London: Department of Health; 1996.
25. Office of National Statistics. Mortality statistics (revised) 34. Rutten-van Molken MP, Postma MJ, Joore MA, Van
1994, England and Wales. London: Her Majesty’s Stationery Genugten ML, Leidl R, Jager JC. Current and future medical
Office; 1996. costs of asthma and chronic obstructive pulmonary disease
26. World Health Organization. World health statistics annual in the Netherlands. Respir Med 1999; 93:779-87.
1995. Geneva: World Health Organization; 1995. 35. Jacobson L, Hertzman P, Lofdahl C-G, Skoogh B-E, Lindgren
27. World Health Organization, Geneva. Available from: URL: B. The economic impact of asthma and COPD in Sweden
www.who.int 1980 and 1991. Respir Med 2000; 94:247-55.
28. Renzetti AD, McClement JH, Litt BD. The Veterans 36. Fauroux B, Howard P, Muir JF. Home treatment for chronic
Administration Cooperative Study of Pulmonary Function. III: respiratory insufficiency: the situation in Europe in 1992. The
Mortality in relation to respiratory function in chronic obstruc- European Working Group on Home Treatment for Chronic
tive pulmonary disease. Am J Med 1966; 41:115-29. Respiratory Insufficiency. Eur Respir J 1994; 7:1721-6.

18 BURDEN OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 19

CHAPTER

RISK FACTORS
GOLD_WR_05 8/18/05 12:56 PM Page 20

CHAPTER 3: RISK FACTORS


example, gender may influence whether a person takes
up smoking or experiences certain occupational or envi-
KEY POINTS: ronmental exposures; socioeconomic status may be
• Risk factors for COPD include both host factors linked to a child's birth weight; longer life expectancy will
and environmental exposures, and the disease allow greater lifetime exposure to risk factors; etc.
usually arises from an interaction between these Understanding the relationships and interactions among
two types of factors. risk factors is a crucial area of ongoing investigation.

• The host factor that is best documented is a rare


Figure 3-1. Risk Factors for COPD
hereditary deficiency of alpha-1 antitrypsin. Other
genes involved in the pathogenesis of COPD Host Factors • Genes (e.g., alpha-1
have not yet been identified. antitrypsin deficiency)
• Airway Hyperresponsiveness
• The major environmental factors are tobacco • Lung Growth
smoke, occupational dusts and chemicals
(vapors, irritants, fumes), and indoor/outdoor air Exposures • Tobacco Smoke
• Occupational Dusts and
pollution.
Chemicals
• Indoor and Outdoor Air Pollution
• Infections
INTRODUCTION • Socioeconomic Status

The identification of risk factors is an important step


toward developing strategies for prevention and treatment The best-documented host factor is a severe hereditary
of any disease. Identification of cigarette smoking as an deficiency of alpha-1 antitrypsin. The major environmental
important risk factor for COPD has led to the incorporation factors are tobacco smoke, occupational dusts and
of smoking cessation programs as a key element of chemicals (vapors, irritants, fumes), and indoor and outdoor
COPD prevention, as well as an important intervention air pollution. However, it is very difficult to demonstrate that
for patients who already have the disease. However, a given risk factor is sufficient to cause the disease.
although smoking is the best-studied COPD risk factor, it
is not the only one. Further studies of other risk factors Data are not available to determine whether the increasing
could lead to similar powerful interventions. prevalence of respiratory symptoms and the accelerated
rate of lung function decline that occur with age reflect
Much of the evidence concerning risk factors for COPD the cumulative exposure to respiratory particles, irritants,
comes from cross-sectional epidemiological studies that fumes, vapors, etc., or host-related phenomena such as
identify associations rather than cause-and-effect rela- the loss of elastic recoil of lung tissue and stiffening of
tionships. Although several longitudinal studies (which the chest wall. The field of normal lung aging has been
are capable of revealing causal relationships) of COPD only minimally explored and more work is required.
have followed groups and populations for up to 20 years,
none of them has monitored the progression of the disease The role of gender as a risk factor for COPD remains
through its entire course. Thus, current understanding of unclear. In the past, most studies showed that COPD
risk factors for COPD is in many respects incomplete. prevalence and mortality were greater among men than
women1-4. More recent studies5,6 from developed countries
Figure 3-1 provides a summary of risk factors for COPD. show that the prevalence of the disease is almost equal
The division into "Host Factors" and "Exposures" reflects in men and women, which probably reflects changing
the current understanding of COPD as resulting from an patterns of tobacco smoking. Some studies have in fact
interaction between the two types of factors. Thus, of two suggested that women are more susceptible to the
people with the same smoking history, only one may effects of tobacco smoke than men4,7. This is an important
develop COPD due to differences in genetic predisposition question given the increasing rate of smoking among
to the disease, or in how long they live. Risk factors for women in both developed and developing countries.
COPD may also be related in more complex ways. For The role of nutritional status as an independent risk factor

20 RISK FACTORS
GOLD_WR_05 8/18/05 12:56 PM Page 21

for the development of COPD is unclear. Malnutrition and airway hyperresponsiveness and increased risk of
weight loss can reduce respiratory muscle strength and developing COPD was originally described by Orie and
endurance, apparently by reducing both respiratory muscle colleagues23 and termed the "Dutch hypothesis."
mass and the strength of the remaining muscle fibers8. Asthmatics, as a group, experience a slightly accelerated
The association of starvation and anabolic/catabolic loss of lung function24,25 compared to non-asthmatics, as
status with the development of emphysema has been do smokers with airway hyperresponsiveness compared
shown in experimental studies in animals9. to normal smokers26. How these trends are related to the
development of COPD is unknown, however. Airway
hyperresponsiveness may also develop after exposure to
HOST FACTORS tobacco smoke or other environmental insults and thus
may be a result of smoking-related airway disease.
Genes
Lung Growth
It is believed that many genetic factors increase (or
decrease) a person's risk of developing COPD. Studies Lung growth is related to processes occurring during
have demonstrated an increased risk of COPD within gestation, birth weight, and exposures during childhood27-31.
families with COPD probands. Some of this risk may be Reduced maximal attained lung function (as measured by
due to shared environmental factors, but several studies spirometry) may identify individuals who are at increased
in diverse populations also suggest a shared genetic risk10,11. risk for the development of COPD32.

The genetic risk factor that is best documented is a


severe hereditary deficiency of alpha-1 antitrypsin12-14, a EXPOSURES
major circulating inhibitor of serine proteases. This rare
hereditary deficiency is a recessive trait most commonly It may be helpful conceptually to think of a person's
seen in individuals of Northern European origin. exposures in terms of his or her total burden of inhaled
Premature and accelerated development of panlobular particles (Figure 3-2). Each type of particle, depending
emphysema and decline in lung function occur in both on its size and composition, may contribute a different
smokers and nonsmokers with the severe deficiency, weight to the risk, and the total risk will depend on the
although smoking increases the risk appreciably. There integral of the inhaled exposures. Of the many inhalational
is considerable variation between individuals in the exposures that people may encounter over a lifetime, only
extent and severity of the emphysema and the rate of tobacco smoke2,33-39 and occupational dusts and chemicals
lung function decline. Although alpha-1 antitrypsin (vapors, irritants, and fumes)40,41 are known to cause
deficiency is relevant to only a small part of the world's COPD on their own. Tobacco smoke and occupational
population, it illustrates the interaction between host exposures also appear to act additively to increase a
factors and environmental exposures leading to COPD. person's risk of developing COPD.
In this way, it provides a model for how other genetic risk
factors are thought to contribute to COPD. Tobacco Smoke
Exploratory studies have revealed a number of candidate Cigarette smoking is by far the most important risk factor
genes that may influence a person's risk of COPD, for COPD and the most important way that tobacco
including ABO secretor status15,16, microsomal epoxide contributes to the risk of COPD. Cigarette smokers have
hydrolase17, glutathione S-transferase18, alpha-1 antichy- a higher prevalence of respiratory symptoms and lung
motrypsin19, the complement component GcG20, cytokine function abnormalities, a greater annual rate of decline in
TNF-21, and microsatellite instability22. However, when FEV1, and a greater COPD mortality rate than nonsmokers.
several studies of a given trait are available, the results These differences between cigarette smokers and non-
are often inconsistent. Several of these genes are smokers increase in direct proportion to the quantity of
thought to be involved in inflammation, and therefore are smoking. Pipe and cigar smokers have greater COPD
related to potential pathogenic mechanisms of COPD. morbidity and mortality rates than nonsmokers, although
their rates are lower than those for cigarette smokers33.
Airway Hyperresponsiveness Other types of tobacco smoking popular in various
countries are also risk factors for COPD, although their
Asthma and airway hyperresponsiveness, identified as risk relative to cigarette smoking has not been reported.
risk factors that contribute to the development of COPD,
are complex disorders related to a number of genetic and Age at starting to smoke, total pack-years smoked, and
environmental factors. The relationship between asthma/ current smoking status are predictive of COPD mortality.

RISK FACTORS 21
GOLD_WR_05 8/18/05 12:56 PM Page 22

Figure 3-2. Total Burden of Inhaled Particles Figure 3-3. Interaction of Smoking
and Occupational Exposures41

Cigarette Smoke
Occupational Dusts
& Chemicals
Environmental Tobacco
Smoke (ETS)
Indoor/Outdoor
Air Pollution

Excerpted with permission from Kaufmann F, Drouet D, Lellouch J, Brille D. International


Journal of Epidemiology 1979; 8:201-12. Copyright 1979 Oxford University Press.
Not all smokers develop clinically significant COPD,
which suggests that genetic factors must modify each
individual's risk. Although it is unclear what percentage and gas or fume exposure may have an additive effect
of smokers develop the disease, the commonly cited on the risk of COPD48-50.
figure of 15-20% is likely an underestimate because
COPD is both underdiagnosed and underappreciated. Indoor and Outdoor Air Pollution

Passive exposure to cigarette smoke (also known as High levels of urban air pollution are harmful to individuals
environmental tobacco smoke or ETS) may also contribute with existing heart or lung disease. The role of outdoor
to respiratory symptoms and COPD by increasing the air pollution in causing COPD is unclear, but appears to
lungs' total burden of inhaled particles and gases2,42,43. be small when compared with that of cigarette smoking.
Smoking during pregnancy may also pose a risk for the The relative effect of short-term, high peak exposures
fetus, by affecting lung growth and development in utero and long-term, low-level exposures is a question yet to
and possibly the priming of the immune system32,44. be resolved.

Occupational Dusts and Chemicals Over the past two decades, air pollution in most cities in
developed countries has decreased appreciably. In
Occupational dusts and chemicals (vapors, irritants, contrast, air pollution has increased markedly in many
and fumes) can also cause COPD when the exposures cities in developing countries. Although it is not clear
are sufficiently intense or prolonged, such as those which specific elements of ambient air pollution are
experienced by miners in many countries. These harmful, there is some evidence that particles found in
exposures can both cause COPD independently of polluted air will add to a person's total inhaled burden.
cigarette smoking and increase the risk in the presence Indoor air pollution from biomass fuel has been implicated
of concurrent cigarette smoking (Figure 3-3)41. Exposure as a risk factor for the development of COPD. This exposure
to coal dust alone in sufficient doses can produce airflow is greatest in regions where biomass fuel is used for
limitation45,46. cooking and heating in poorly vented dwellings, leading
to high levels of particulate matter in indoor air51-61.
Exposure to particulate matter, irritants, organic dusts,
and sensitizing agents can cause an increase in airway Infections
hyperresponsiveness47, especially in airways already
damaged by other occupational exposures, cigarette A history of severe childhood infection has been associat-
smoke, or asthma. There is some evidence from ed with reduced lung function and increased respiratory
community studies that a combination of dust exposure symptoms in adulthood32. There are several possible

22 RISK FACTORS
GOLD_WR_05 8/18/05 12:56 PM Page 23

explanations for this association (which are not mutually 9. Sahebjami H, Vassallo CL. Influence of starvation on
exclusive). There may be an increased diagnosis of enzyme-induced emphysema. J Appl Physiol 1980; 48:284-8.
severe infections in children who have underlying airway 10. Silverman EK, Speizer FE. Risk factors for the develop-
hyperresponsiveness, itself considered a risk factor for ment of chronic obstructive pulmonary disease. Med Clin
COPD. Viral infections may be related to another factor, North Am 1996; 80:501-22.
such as birth weight, that is related to COPD. 11. Chen Y. Genetics and pulmonary medicine. 10: Genetic
epidemiology of pulmonary function. Thorax 1999; 54:818-24.
HIV infection has been shown to accelerate the onset of 12. Laurell CB, Eriksson S. The electrophoretic alpha-1 globu-
smoking-induced emphysema; HIV-induced pulmonary lin pattern of serum in alpha-1 antitrypsin deficiency. Scand
inflammation may play a role in this process62-66. J Clin Lab Invest 1963; 15:132-40.
13. Hubbard RC, Crystal RG. Antiproteases. In: Crystal RB,
Socioeconomic Status West JB, Barnes PJ, Cherniack NS, Weibel ER, eds. The
lung: scientific foundations. New York: Raven Press; 1991.
There is evidence that the risk of developing COPD is p. 1775-87.
inversely related to socioeconomic status65. It is not clear, 14. McElvaney NG, Crystal RG. Inherited susceptibility of the
however, whether this pattern reflects exposures to lung to proteolytic injury. In: Crystal RG, West JB, Weibel
indoor and outdoor air pollutants, crowding, poor nutrition, ER, Barnes PJ, eds. The lung: scientific foundations.
or other factors that are related to low socioeconomic Philadelphia: Lippincott-Raven; 1997. p. 2537-53.
status60,66. 15. Khoury MJ, Beaty TH, Newill CA, Bryant S, Cohen BH.
Genetic-environmental interactions in chronic airways
REFERENCES obstruction. Int J Epidemiol 1986; 15:65-72.
16. Cohen BH, Bias WB, Chase GA, Diamond EL, Graves CG,
1. Feinleib M, Rosenberg HM, Collins JG, Delozier JE, Levy DA, et al. Is ABH nonsecretor status a risk factor for
Pokras R, Chevarley FM. Trends in COPD morbidity and obstructive lung disease? Am J Epidemiol 1980; 111:285-91.
mortality in the United States. Am Rev Respir Dis 1989; 17. Smith CA, Harrison DJ. Association between polymor-
140:S9-18. phism in gene for microsomal epoxide hydrolase and sus-
2. Buist AS, Vollmer WM. Smoking and other risk factors. In: ceptibility to emphysema. Lancet 1997; 350:630-3.
Murray JF, Nadel JA, eds. Textbook of respiratory medi- 18. Harrison DJ, Cantlay AM, Rae F, Lamb D, Smith CA.
cine. Philadelphia: WB Saunders; 1994. p. 1259-87. Frequency of glutathione S-transferase M1 deletion in
3. Thom TJ. International comparisons in COPD mortality. Am smokers with emphysema and lung cancer. Hum Exp
Rev Respir Dis 1989; 140:S27-34. Toxicol 1997; 16:356-60.
4. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking, 19. Faber JP, Poller W, Olek K, Baumann U, Carlson J,
changes in smoking habits, and rate of decline in FEV1: Lindmark B, et al. The molecular basis of alpha 1-antichy-
new insight into gender differences. Eur Respir J 1994; motrypsin deficiency in a heterozygote with liver and lung
7:1056-61. disease. J Hepatol 1993; 18:313-21.
5. National Center for Health Statistics. Current estimates 20. Schellenberg D, Pare PD, Weir TD, Spinelli JJ, Walker BA,
from the National Health Interview Survey, United States, Sandford AJ. Vitamin D binding protein variants and the risk
1995. Washington, DC: Department of Health and Human of COPD. Am J Respir Crit Care Med 1998; 157:957-61.
Services, Public Health Service, Vital and Health Statistics; 21. Huang SL, Su CH, Chang SC. Tumor necrosis factor-alpha
1995. Publication No. 96-1527. gene polymorphism in chronic bronchitis. Am J Respir Crit
6. National Heart, Lung, and Blood Institute. Morbidity & Care Med 1997; 156:1436-9.
mortality: chartbook on cardiovascular, lung, and blood 22. Siafakas NM, Tzortzaki EG, Sourvinos G, Bouros D,
diseases. Bethesda, MD: US Department of Health and Tzanakis N, Kafatos A, et al. Microsatellite DNA instability in
Human Services, Public Health Service, National COPD. Chest 1999; 116:47-51.
Institutes of Health; 1998. Available from: 23. Orie NGM, Sluiter HJ, De Vreis K, Tammerling K, Wikop J.
URL: www.nhlbi.nih.gov/nhlbi/seiin/other/cht-book/html The host factor in bronchitis. In: Orie NGM, Sluiter HJ, eds.
7. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey Bronchitis, an international symposium. Assen, Netherlands:
WC, Buist AS, et al. Effects of smoking intervention and Royal Vangorcum; 1961. p. 43-59.
the use of an inhaled anticholinergic bronchodilator on the 24. Peat JK, Woolcock AJ, Cullen K. Rate of decline of lung
rate of decline of FEV1. The Lung Health Study. JAMA function in subjects with asthma. Eur J Respir Dis 1987;
1994; 272:1497-505. 70:171-9.
8. Wilson DO, Rogers RM, Wright EC, Anthonisen NR. Body 25. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-
weight in chronic obstructive pulmonary disease. The year follow-up study of ventilatory function in adults with
National Institutes of Health Intermittent Positive-Pressure asthma. N Engl J Med 1998; 339:1194-200.
Breathing Trial. Am Rev Respir Dis 1989; 139:1435-8.

RISK FACTORS 23
GOLD_WR_05 8/18/05 12:56 PM Page 24

26. Tashkin DP, Altose MD, Connett JE, Kanner RE, Lee WW, 42. Leuenberger P, Schwartz J, Ackermann-Liebrich U, Blaser
Wise RA. Methacholine reactivity predicts changes in lung K, Bolognini G, Bongard JP, et al. Passive smoking exposure
function over time in smokers with early chronic obstructive in adults and chronic respiratory symptoms (SAPALDIA
pulmonary disease. The Lung Health Study Research Study). Swiss Study on Air Pollution and Lung Diseases in
Group. Am J Respir Crit Care Med 1996; 153:1802-11. Adults, SAPALDIA Team. Am J Respir Crit Care Med 1994;
27. Morgan WJ. Maternal smoking and infant lung function. 150:1222-8.
Further evidence for an in utero effect [editorial; comment]. 43. Dayal HH, Khuder S, Sharrar R, Trieff N. Passive smoking in
Am J Respir Crit Care Med 1998; 158:689-90. obstructive respiratory disease in an industrialized urban
28. Hagstrom B, Nyberg P, Nilsson PM. Asthma in adult life - is population. Environ Res 1994; 65:161-71.
there an association with birth weight? Scand J Prim Health 44. Holt PG. Immune and inflammatory function in cigarette
Care 1998; 16:117-20. smokers. Thorax 1987; 42:241-9.
29. Svanes C, Omenaas E, Heuch JM, Irgens LM, Gulsvik A. 45. US Centers for Disease Control and Prevention. Criteria
Birth characteristics and asthma symptoms in young adults: for a recommended standard: occupational exposure to
results from a population-based cohort study in Norway. Eur respirable coal mine dust. Morgantown, WV: National
Respir J 1998; 12:1366-70. Institute of Occupational Safety and Health; 1995.
30. Todisco T, de Benedictis FM, Iannacci L, Baglioni S, Eslami Publication No. 95-106.
A, Todisco E, et al. Mild prematurity and respiratory func- 46. Heppleston AG. Prevalence and pathogenesis of pneumoco-
tions. Eur J Pediatr 1993; 152:55-8. niosis in coal workers. Environ Health Perspect 1988;
31. Stein CE, Kumaran K, Fall CH, Shaheen SO, Osmond C, 78:159-70.
Barker DJ. Relation of fetal growth to adult lung function in 47. Niewoehner DE. Anatomic and pathophysiological correla-
South India. Thorax 1997; 52:895-9. tions in COPD. In: Baum GL, Crapo JD, Celli BR, Karlinsky
32. Tager IB, Segal MR, Speizer FE, Weiss ST. The natural his- JB, eds. Textbook of pulmonary diseases. Philadelphia:
tory of forced expiratory volumes. Effect of cigarette smoking Lippincott-Raven; 1998. p. 823-42.
and respiratory symptoms. Am Rev Respir Dis 1988; 48. Bakke S, Baste V, Hanoa R, Gulsvik A. Prevalence of
138:837-49. obstructive lung disease in a general population: relation to
33. US Surgeon General. The health consequences of smoking: occupational title and exposure to some airborne agents.
chronic obstructive pulmonary disease. Washington, DC: Thorax 1991; 46:863-70.
US Department of Health and Human Services; 1984. 49. Humerfelt S, Gulsvik A, Skjaerven R, Nilssen S, Kvale G,
Publication No. 84-50205. Sulheim O, et al. Decline in FEV1 and airflow limitation relat-
34. Higgins MW, Thom T. Incidence, prevalence, and mortality: ed to occupational exposures in men of an urban communi-
intra- and inter-country differences. In: Hensley M, Saunders ty. Eur Respir J 1993; 6:1095-103.
N, eds. Clinical epidemiology of chronic obstructive pul- 50. Humerfelt S, Eide GE, Gulsvik A. Association of years of
monary disease. New York: Marcel Dekker; 1989. p. 23-43. occupational quartz exposure with spirometric airflow limita-
35. Sherrill DL, Lebowitz MD, Burrows B. Epidemiology of tion in Norwegian men aged 30-46 years. Thorax 1998;
chronic obstructive pulmonary disease. Clin Chest Med 53:649-55.
1990; 11:375-87. 51. Chen JC, Mannino MD. Worldwide epidemiology of chronic
36. Auerbach O, Hammond EC, Garfinkel L, Benante C. Relation obstructive pulmonary disease. Current Opinion in
of smoking and age to emphysema. Whole-lung section Pulmonary Medicine 1999; 5:93-9.
study. N Engl J Med 1972; 286:853-7. 52. Perez-Padilla R, Regalado U, Vedal S, Pare P, Chapela R,
37. Burrows B, Knudson RJ, Cline MG, Lebowitz MD. Sansores R, et al. Exposure to biomass smoke and chronic
Quantitative relationships between cigarette smoking and airway disease in Mexican women. Am J Respir Crit Care
ventilatory function. Am Rev Respir Dis 1977; 115:195-205. Med 1996; 154:701-6.
38. Lebowitz MD, Burrows B. Quantitative relationships between 53. Dossing M, Khan J, al-Rabiah F. Risk factors for chronic
cigarette smoking and chronic productive cough. Int J obstructive lung disease in Saudi Arabia. Respiratory Med
Epidemiol 1977; 6:107-13. 1994; 88:519-22.
39. Higgins MW, Keller JB, Becker M, Howatt W, Landis JR, 54. Behera D, Jindal SK. Respiratory symptoms in Indian
Rotman H, et al. An index of risk for obstructive airways dis- women using domestic cooking fuels. Chest 1991; 100:385-8.
ease. Am Rev Respir Dis 1982; 125:144-51. 55. Amoli K. Bronchopulmonary disease in Iranian housewives
40. Becklake MR. Occupational exposures: evidence for a chronically exposed to indoor smoke. Eur Respir J 1998;
causal association with chronic obstructive pulmonary dis- 11:659-63.
ease. Am Rev Respir Dis 1989; 140:S85-91. 56. Dennis R, Maldonado D, Norman S, Baena E, Martinez G.
41. Kauffmann F, Drouet D, Lellouch J, Brille D. Twelve years Woodsmoke exposure and risk for obstructive airways dis-
spirometric changes among Paris area workers. Int J ease among women. Chest 1996; 109:115-9.
Epidemiol 1979; 8:201-12. 57. Pandey MR. Prevalence of chronic bronchitis in a rural com-
munity of the Hill Region of Nepal. Thorax 1984; 39:331-6.

24 RISK FACTORS
GOLD_WR_05 8/18/05 12:56 PM Page 25

58. Pandey MR. Domestic smoke pollution and chronic bronchi- 63. Diaz PT, King MA, Pacht ER, Wewers MD, Gadek JE,
tis in a rural community of the Hill Region of Nepal. Thorax Nagaraja HN, et al. Increased susceptibility to pulmonary
1984; 39:337-9. emphysema among HIV-seropositive smokers. Ann Intern
59. Samet JM, Marbury M, Spengler J. Health effects and Med 2000; 132:369-72.
sources of indoor air pollution. Am Rev Respir Dis 1987; 64. Diaz PT, King MA, Pacht ER, Wewers MD, Gadek JE, Neal
136:1486-508. D, et al. The pathophysiology of pulmonary diffusion impair-
60. Tao X, Hong CJ, Yu S, Chen B, Zhu H, Yang M. Priority ment in human immunodeficiency virus infection. Am J
among air pollution factors for preventing chronic obstructive Respir Crit Care Med 1999; 160:272-7.
pulmonary disease in Shanghai. Sci Total Environ 1992; 65. Prescott E, Lange P, Vestbo J. Socioeconomic status, lung
127:57-67. function and admission to hospital for COPD: results from
61. Smith KR. National burden of disease in India from indoor the Copenhagen City Heart Study. Eur Respir J 1999;
air pollution. Proc Natl Acad Sci USA 2000; 97:13286-93. 13:1109-14.
62. Diaz PT, Clanton TL, Pacht ER. Emphysema-like pulmonary 66. Strachan DP. Epidemiology: a British perspective. In:
disease associated with human immunodeficiency virus Calverley PMA, Pride NB, eds. Chronic obstructive pulmonary
infection. Ann Intern Med 1992; 116:124-8. disease. London: Chapman and Hall; 1995. p. 47-67.

RISK FACTORS 25
GOLD_WR_05 8/18/05 12:56 PM Page 26

26 RISK FACTORS
GOLD_WR_05 8/18/05 12:56 PM Page 27

CHAPTER

PATHOGENESIS,
PATHOLOGY, AND
PATHOPHYSIOLOGY
GOLD_WR_05 8/18/05 12:56 PM Page 28

CHAPTER 4: PATHOGENESIS, PATHOLOGY,


AND PATHOPHYSIOLOGY
KEY POINTS: • In advanced COPD, peripheral airways obstruction,
parenchymal destruction, and pulmonary vascular
• Exposure to inhaled noxious particles and gases abnormalities reduce the lung's capacity for gas
causes inflammation of the lungs that can lead to
exchange, producing hypoxemia and, later on,
COPD if the normal protective and/or repair
hypercapnia. Inequality in the ventilation/perfusion
mechanisms are overwhelmed or defective.
ratio (VA/Q) is the major mechanism behind
hypoxemia in COPD.
• Exacerbations of COPD are associated with an
increase in airway inflammation.
• Pulmonary hypertension develops late in the
course of COPD. It is the major cardiovascular
• Although inflammation is important in both
complication of COPD and is associated with a
diseases, the inflammatory response in COPD is
poor prognosis.
markedly different from that in asthma.
• COPD is associated with systemic inflammation
• In addition to inflammation, two other processes and skeletal muscle dysfunction that may
thought to be important in the pathogenesis of
contribute to limitation of exercise capacity and
COPD are an imbalance of proteinases and
decline of health status.
antiproteinases in the lung, and oxidative stress.

• Pathological changes characteristic of COPD are INTRODUCTION


found in the central airways, peripheral airways,
lung parenchyma, and pulmonary vasculature. Inhaled noxious particles and gases that lead to COPD
cause lung inflammation, induce tissue destruction,
• The peripheral airways become the major site of impair the defense mechanisms that serve to limit the
airways obstruction in COPD. The structural destruction, and disrupt the repair mechanisms that may
changes in the airway wall are the most important be able to restore tissue structure in the face of some
cause of the increase in peripheral airways injuries. The results of lung tissue damage are mucus
resistance in COPD. Inflammatory changes such hypersecretion, airway narrowing and fibrosis, destruction
as airway edema and mucus hypersecretion also of the parenchyma (emphysema), and vascular changes.
contribute to airway narrowing. In turn, these pathological changes lead to airflow limitation
and the other physiological abnormalities characteristic of
• Most common in COPD patients is the COPD.
centrilobular form of emphysema, which involves
dilatation and destruction of the respiratory Much of the information concerning the pathogenesis
bronchioles. of COPD comes from studies in experimental animals or
in vitro systems. These experimental systems are limited
• Physiological changes characteristic of the as they differ from human disease in a number of
disease include mucus hypersecretion, ciliary respects. Studies in human subjects of the pathogenesis,
pathology, and pathophysiology of COPD are often limited
dysfunction, airflow limitation, pulmonary hyperin-
by patient selection, small numbers of subjects, and limited
flation, gas exchange abnormalities, pulmonary
access to the relevant tissue. Therefore, an evidence-
hypertension, and cor pulmonale, and they
based perspective on these topics is in many respects
usually develop in this order over the course of
incomplete.
the disease.
PATHOGENESIS
• The irreversible component of airflow limitation is
primarily due to remodeling of the small airways. COPD is characterized by chronic inflammation through-
Parenchymal destruction (emphysema) also out the airways, parenchyma, and pulmonary vasculature.
contributes but plays a smaller role. The intensity and cellular and molecular characteristics of
the inflammation vary as the disease progresses. Over

28 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 29

time, inflammation damages the lungs and leads to the Inflammatory Cells
pathologic changes characteristic of COPD.
COPD is characterized by an increase in neutrophils,
In addition to inflammation, two other processes thought macrophages, and T lymphocytes (especially CD8+) in
to be important in the pathogenesis of COPD are an various parts of the lung (Figure 4-3). There may also be
imbalance of proteinases and antiproteinases in the lung, an increase in eosinophils in some patients, particularly
and oxidative stress. These processes may themselves during exacerbations. These increases are brought about
be consequences of inflammation, or they may arise from by increases in inflammatory cell recruitment, survival,
environmental (e.g., oxidant compounds in cigarette and/or activation. Many studies reveal a correlation
smoke) or genetic (e.g., alpha-1 antitrypsin deficiency) between the number of inflammatory cells of various
factors. Figure 4-1 details the interactions between these types in the lung and the severity of COPD1-10.
mechanisms. The multiplicity of cells and mediators
thought to be involved in the pathogenesis of COPD is
presented schematically in Figure 4-2. Figure 4-3. Sites of Inflammatory
Cell Increases in COPD

Figure 4-1. Pathogenesis of COPD


Large Airways • Macrophages
• T lymphocytes (especially CD8+)
• Neutrophils (severe disease only)
Noxious particles • Eosinophils (in some patients)
and gases
Small Airways • Macrophages
• T lymphocytes (especially CD8+)
• Eosinophils (in some patients)
Lung Inflammation
Anti-oxidants Antiproteinases
Parenchyma • Macrophages
• T lymphocytes (especially CD8+)
Oxidative stress Proteinases • Neutrophils

Pulmonary Arteries • T lymphocytes (especially CD8+)


Repair mechanisms
• Neutrophils
COPD pathology

Neutrophils. Increased numbers of activated neutrophils


are found in sputum and bronchoalveolar lavage (BAL)
fluid of patients with COPD4,5,8,9, although the role of
Figure 4-2. Cells and Mediators Involved neutrophils in COPD is not yet clear. Neutrophils are
in the Pathogenesis of COPD also increased in smokers without COPD11. However,
neutrophils are little increased in airway and parenchyma
tissue sections, which may reflect their rapid transit
MEDIATORS
LTB4
through these parts of the lung. Induced sputum studies
IL-8, GRO-1 also show an increase in myeloperoxidase (MPO)
MCP-1, MIP-1 and human neutrophil lipocalin, indicating neutrophil
CELLS GM-CSF 4 activation12. Exacerbations of COPD are characterized by
EFFECTS
Macrophages Endothelin a marked increase in the number of neutrophils in BAL
Mucus hypersecretion
Neutrophils Substance P fluid13. Neutrophils secrete several proteinases, including
Fibrosis
CD8+ lymphocytes -1 Alveolar wall neutrophil elastase (NE), neutrophil cathepsin G, and
Eosinophils PROTEINASES destruction
Epithelial cells Neutrophil elastase
neutrophil proteinase-3, which may contribute to
Fibroblasts Cathepsins parenchymal destruction and chronic mucus hypersecretion.
Proteinase-3
MMPs Macrophages. Increased numbers of macrophages are
present in the large and small airways and lung
Printed with permission of Dr. Peter J. Barnes.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 29


GOLD_WR_05 8/18/05 12:56 PM Page 30

parenchyma of patients with COPD, as reflected in that some form of down-regulation of inflammatory
histopathology, BAL, bronchial biopsy, and induced mediator release may occur in epithelial cells of indi-
sputum studies2,4-6,9. In patients with emphysema, viduals with COPD20.
macrophages are localized to sites of alveolar wall
destruction1. Macrophages likely play an orchestrating Inflammatory Mediators
role in COPD inflammation by releasing mediators
such as tumor necrosis factor- (TNF-), interleukin Activated inflammatory cells in COPD release a variety
8 (IL-8), and leukotriene B4 (LTB4), which promote of mediators, including a spectrum of potent proteinases22,23,
neutrophilic inflammation. oxidants24, and toxic peptides25. Many of the mediators
thought to be important in the disease – notably LTB426,
T lymphocytes. Histopathology and bronchial biopsy IL-84,27, and TNF- 4,16 – are capable of damaging lung
studies show an increase in T lymphocytes, especially structures and/or sustaining neutrophilic inflammation.
CD8+ (cytotoxic) cells, throughout the lungs of patients The damage induced by these moieties may further
with COPD1,2,10,14. Their role in COPD inflammation is potentiate inflammation by releasing chemotactic
not yet fully understood, but one way that CD8+ cells peptides from the extracellular matrix28. Little is yet
may contribute to COPD is by releasing perforin, known about the specific role of these inflammatory
granzyme-B, and TNF-, which can cause the cytolysis mediators in COPD. Studies of the therapeutic use of
and apoptosis of alveolar epithelial cells15 that may be selective mediator antagonists should identify the
responsible for the persistence of inflammation. An molecules relevant in COPD.
increased number of lymphocyte-like natural killer (NK)
Leukotriene B4 (LTB4). LTB4, a potent chemoattractant
cells has also been reported in patients with severe
of neutrophils, is found at increased levels in the sputum
COPD3.
of patients with COPD26. It is probably derived from
alveolar macrophages, which secrete more LTB4 in
Eosinophils. The presence and role of eosinophils
patients with COPD. Several potent LTB4 receptor
in COPD are uncertain. Some bronchial biopsy
antagonists have been developed for clinical studies
studies show eosinophils increased in the airways of
and should elucidate further the role of this mediator
some patients with stable COPD6,16. However, some
in COPD. So far there is no evidence that cysteinyl
of these patients may have had coexisting asthma, as
leukotrienes (LTC4, LTD4, LTE4) are involved in
other studies report no increase in eosinophils in
COPD. Selective antagonists of the cysteinyl
COPD patients2. The levels of eosinophil cationic
leukotriene 1 receptor (CysLT1) have proven helpful in
protein (ECP) and eosinophil peroxidase (EPO) in
patients with asthma and studies of these drugs in
induced sputum are elevated in COPD, suggesting
COPD patients are now underway. The role of the
that eosinophils may be present but degranulated, and
cysteinyl leukotriene 2 receptor (CysLT2) in respiratory
therefore no longer recognizable by light microscopy12.
disease is as yet unknown29.
The high levels of neutrophil elastase (NE) often found
in COPD may be responsible for this degranulation17. Interleukin 8 (IL-8). IL-8, a selective chemoattractant
Most studies agree that airway eosinophils are of neutrophils that may be secreted by macrophages,
increased during exacerbations of COPD18,19. neutrophils, and airway epithelial cells, is present at
high concentrations in induced sputum and BAL
Epithelial cells. Airway and alveolar epithelial cells fluid of patients with COPD4,27. IL-8 may play a
are likely to be important sources of inflammatory primary role in the activation of both neutrophils and
mediators in COPD, though their role in inflammation eosinophils in the airways of COPD patients and may
in this disease has not yet been thoroughly studied. serve as a marker in evaluating the severity of airway
Exposure of nasal or bronchial epithelial cells from inflammation27.
healthy volunteers to nitrogen dioxide (NO2), ozone
(O3), and diesel exhaust particles results in significant Tumor necrosis factor- (TNF-). TNF- activates
synthesis and release of proinflammatory mediators, the transcription factor nuclear factor-B (NF-B),
including eicosanoids, cytokines, and adhesion which in turn activates the IL-8 gene in epithelial cells
molecules20. The adhesion molecule E-selectin, and macrophages (Figure 4-4). TNF- is present at
involved in recruitment and adhesion of neutrophils, is high concentrations in sputum4 and is detectable in
up-regulated on airway epithelial cells in COPD patients21. bronchial biopsies16 in patients with COPD. TNF-
Cultured human bronchial epithelial cells from COPD serum levels and production by peripheral blood
patients release lower levels of inflammatory mediators monocytes are increased in weight-losing COPD
such as TNF- and IL-8 than similar preparations from patients, suggesting that this mediator may play a role in
nonsmokers or smokers without COPD, suggesting the cachexia of severe COPD30.

30 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 31

Figure 4-4. Interaction Between • Transforming growth factor-ß (TGF-ß) and epidermal
Macrophages, Neutrophils, and Epithelial Cells growth factor (EGF) show increased expression in
epithelial cells and submucosal cells (eosinophils and
fibroblasts) in COPD patients33. These mediators
may play a role in airway remodeling (fibrosis and
narrowing) in COPD34.

• Endothelin-1 (ET-1), a potent endothelium-derived


vasoconstrictor peptide, is found at increased
concentrations in induced sputum of patients with
COPD35. Patients with severe COPD also have
elevated plasma levels of ET-1, which is probably
related to their chronic hypoxemia36.

• Neuropeptides, such as substance P, calcitonin


gene-related peptide, and vasoactive intestinal
Cigarette smoke activates macrophages and epithelial peptide (VIP), have potent effects on vascular function
cells to produce tumor necrosis factor- (TNF-), and mucus secretion. An increased concentration
switching on the gene for interleukin-8 (IL-8), which of substance P is found in sputum of patients with
chronic bronchitis37. One bronchial biopsy study
recruits and activates neutrophils. This process occurs
showed an increase in VIP-immunoreactive nerves
via the activation of the transcription factor nuclear fac-
in the vicinity of submucosal glands in patients with
tor-B (NF-B). chronic bronchitis, suggesting that this substance
Printed with permission of Dr. Peter J. Barnes.
may play a role in mucus hypersecretion38. However,
another study showed no significant differences in
Others. Other inflammatory mediators that may be the number of nerves immunoreactive for substance
involved in COPD include the following: P, calcitonin gene-related peptide, or VIP between
COPD patients and healthy subjects39.
• Macrophage chemotactic protein-1 (MCP-1), a
potent chemoattractant of monocytes, is increased • Complement. Activation of the complement pathway
in the BAL fluid of patients with COPD and smokers via generation of the potent chemotaxin C5a may
without COPD, but not in ex-smokers or nonsmokers31. play a significant role in the neutrophil accumulation
Thus, MCP-1 may be involved in macrophage seen in the lungs of patients with COPD40.
recruitment into the lungs in smokers.
Differences Between Inflammation
• Macrophage inflammatory protein-1ß (MIP-1ß) is in COPD and Asthma
increased in the BAL fluid of patients with COPD
compared to smokers, ex-smokers, and nonsmokers31. Although inflammation is important in both diseases,
Macrophage inflammatory protein-1 (MIP-1) the inflammatory response in COPD is markedly different
shows increased expression in airway epithelial cells from that in asthma, as summarized in Figure 4-5.
from COPD patients3 compared to control smokers. However, some patients with COPD also have asthma,
and the inflammation in their lungs may show charac-
• Granulocyte-macrophage colony stimulating factor teristics of both diseases.
(GM-CSF) is found at increased concentrations in
the BAL fluid of patients with stable COPD and at Since inflammation is a feature of COPD, it follows that
markedly elevated levels during exacerbations13. The anti-inflammatory therapies may have clinical benefit in
controlling symptoms, preventing exacerbations, and
number of GM-CSF-immunoreactive macrophages is
slowing the progression of the disease. However, the
also increased in sputum of patients with COPD32.
inflammatory response in COPD appears to be poorly
GM-CSF is important for neutrophil survival and may
responsive to the glucocorticosteroids that are effective
play a role in enhancing neutrophilic inflammation.
anti-inflammatory medications in asthma.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 31


GOLD_WR_05 8/18/05 12:56 PM Page 32

Figure 4-5. Characteristics of Inflammation in COPD and Asthma

COPD Asthma

Cells • Neutrophils • Eosinophils


• Large increase in macrophages • Small increase in macrophages
• Increase in CD8+ T lymphocytes • Increase in CD4+ Th2 lymphocytes
• Activation of mast cells

Mediators • LTB4 • LTD4


• IL-8 • IL-4, IL-5
• TNF- • (Plus many others)

Consequences • Squamous metaplasia of epithelium • Fragile epithelium


• Parenchymal destruction • Thickening of basement membrane
• Mucus metaplasia • Mucus metaplasia
• Glandular enlargement • Glandular enlargement

Response to • Glucocorticosteroids have little or • Glucocorticosteroids inhibit


Treatment no effect inflammation

Inflammation and COPD Risk Factors A number of studies have demonstrated that a variety of
particulates (e.g., diesel exhaust, grain dust) can initiate
The connection between cigarette smoke and inflammation respiratory tract inflammation57-61. It is likely that indoor air
has been most extensively studied41-52. Cigarette smoke pollution derived from the burning of biomass fuels will
activates macrophages and epithelial cells to produce prove to have similar effects.
TNF- and may also cause macrophages to release
other inflammatory mediators, including IL-8 and LTB453,54. Proteinase-Antiproteinase Imbalance

Inflammation is present in the lungs of smokers without a Laurell and Eriksson observed in 1963 that individuals
diagnosis of COPD. This inflammation is similar to, but with a hereditary deficiency of the serum protein alpha-1
less intense than, the inflammation in the lungs of patients antitrypsin, which inhibits a number of serine proteinases
with COPD. For example, induced sputum studies show such as neutrophil elastase, are at increased risk of
that smokers without COPD have a greater proportion of developing emphysema62. Elastin, the target of neutrophil
neutrophils in their lungs than age-matched nonsmokers, elastase, is a major component of alveolar walls, and
but a smaller proportion than COPD patients4,9. Thus, the elastin fragments may perpetuate inflammation by acting
inflammation characteristic of COPD is thought to represent as potent chemotactic agents for macrophages and
an exaggeration of a normal, protective response to neutrophils. These observations led to the hypothesis
inhalational exposures. that an imbalance between proteinases and endogenous
antiproteinases results in lung destruction.
However, not all smokers develop COPD, and why the
normal, protective inflammatory response becomes an Based on many observations, it now seems clear that an
exaggerated, harmful one in some smokers is poorly imbalance of proteinases and antiproteinases may involve
understood. Presumably the inflammation caused by either increased production or activity of proteinases, or
cigarette smoking interacts with other host or environmental inactivation or reduced production of antiproteinases.
factors to produce the excess decline in lung function Often, the imbalance is a consequence of the inflammation
that results in COPD55. Inflammatory changes are also induced by inhalational exposures. For example,
present in bronchial biopsies in ex-smokers, suggesting macrophages, neutrophils, and airway epithelial cells
that the inflammatory response in COPD may persist even release a combination of proteinases. The imbalance
in the absence of continuous exposure to risk factors56. may also be caused by a decrease of antiproteinase
activity by oxidative stress (itself a consequence of

32 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 33

inflammation), cigarette smoke63,64, and possibly other isoprostane F2-III, which is formed by free radical
COPD risk factors. peroxidation of arachidonic acid and believed to be an in
vivo biomarker of lung oxidative stress, is increased in
The concept has also been expanded to include additional both breath condensates71 and urine72 in COPD patients
proteinases and antiproteinases. While neutrophil elastase compared to healthy controls and is increased even more
is likely to be the major proteinase involved in lung during exacerbations.
destruction in alpha-1 antitrypsin deficiency, it may not
be involved in COPD caused by inhalational exposures. Oxidative stress contributes to COPD in a variety of
Additional proteinases that have been implicated in ways. Oxidants can react with, and damage, a variety of
COPD include neutrophil cathepsin G, neutrophil biological molecules, including proteins, lipids, and nucleic
proteinase-3, cathepsins released from macrophages acids, and this can lead to cell dysfunction or death,
(specifically cathepsins B, L, and S), and various matrix as well as damage to the lung extracellular matrix. In
metalloproteinases (MMPs)65. These proteinases are addition to directly damaging the lung, oxidative stress
capable of degrading elastin and also collagen, another contributes to the proteinase-antiproteinase imbalance
main component of alveolar walls. Some proteinases, both by inactivating antiproteinases (such as alpha-1
such as neutrophil elastase66 and neutrophil proteinase-367, antirypsin and SLPI) and by activating proteinases (such
induce mucus secretion, and neutrophil elastase also as MMPs). Oxidants also promote inflammation, for
produces mucus gland hyperplasia68. Thus, proteinases example by activating the transcription factor NF-B,
may be involved in mucus hypersecretion as well as which orchestrates the expression of multiple inflammatory
parenchymal destruction. Antiproteinases thought to genes thought to be important in COPD such as IL-8
be involved in COPD include, in addition to alpha-1 and TNF-. Finally, oxidative stress may contribute to
anti-trypsin, secretory leukoproteinase inhibitor (SLPI) reversible airway narrowing. H2O2 constricts airway
and tissue inhibitors of MMPs (TIMPs). smooth muscle in vitro and isoprostane F2-III is a potent
constrictor of human airways73.
Oxidative Stress
There is increasing evidence that an oxidant/antioxidant PATHOLOGY
imbalance, in favor of oxidants, occurs in COPD. (The
process is summarized in Figure 4-6.) Markers of Pathological changes characteristic of COPD are found in
oxidative stress have been found in the epithelial lining the central airways, peripheral airways, lung parenchyma,
fluid, breath, and urine of cigarette smokers and patients and pulmonary vasculature74. The various lesions are a
with COPD. For example, hydrogen peroxide (H2O2) result of chronic inflammation in the lung, which in turn is
and nitric oxide (NO) are direct measures of oxidants initiated by the inhalation of noxious particles and gases
generated by cigarette smoking or released from
such as those present in cigarette smoke. The lung has
inflammatory leukocytes and epithelial cells. H2O2 is
natural defense mechanisms and a considerable capacity
increased in the breath of patients with stable COPD and
to repair itself, but the working of these mechanisms may
during exacerbations69, and NO is increased in the breath
be affected by genetic traits (e.g., alpha-1 antitrypsin
during exacerbations of COPD70. A prostaglandin isomer,
deficiency) or exposure to other environmental risk factors
(e.g., infection, atmospheric pollution)75, as well as by the
Figure 4-6. Increased Oxidative Stress in COPD
chronic nature of the inflammation and repeated nature of
ANTIOXIDANTS
the injury.
Glutathione

Antiproteinases
Uric acid, bilirubin
Vitamins C, E
Central Airways
SLPI 1-AT NF-B
The central airways include the trachea, bronchi, and
Proteolysis IL-8 TNF  bronchioles greater than 2-4 mm in internal diameter.
O2 -, H2O2
In patients with chronic bronchitis, an inflammatory
OH, ONOO - Neutrophil
recruitment exudate of fluid and cells infiltrates the epithelium lining
the central airways and associated glands and ducts2,42.
The predominant cells in this inflammatory exudate are
Mucus secretion macrophages and CD8+ T lymphocytes2,76. Chronic
Isoprostanes Plasma leak Bronchoconstriction
inflammation in the central airways is also associated
with an increase in the number (metaplasia) of epithelial
Printed with permission of Dr. Peter J. Barnes. goblet and squamous cells; dysfunction, damage, and/or
loss of cilia; enlarged submucosal mucus-secreting

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 33


GOLD_WR_05 8/18/05 12:56 PM Page 34

glands77; an increase in the amount of smooth muscle Figure 4-8. Pathological Changes
and connective tissue in the airway wall78; degeneration of the Peripheral Airways in COPD
of the airway cartilage79,80; and mucus hypersecretion.
The mechanisms of mucus gland hypertrophy and goblet
cell metaplasia have not yet been identified, but animal
studies81,82 show that irritants including cigarette smoke83
can produce these changes. The various pathological
changes in the central airways (Figure 4-7) are responsible
for the symptoms of chronic cough and sputum production,
which identify people at risk for COPD and may continue
to be present throughout the course of the disease.
Thus, these pathological changes may be present either
on their own or in combination with the changes in the
peripheral airways and lung parenchyma described
below.

Figure 4-7. Pathological Changes


of the Central Airways in COPD

Histological sections of peripheral airways from


patients who are cigarette smokers. (A) is a
nearly normal airway; (B) shows a plug of mucoid
exudate in the lumen with little or no evidence of
inflammation in the wall; (C) shows the presence of
an inflammatory exudate in the wall and lumen of
the airway; and (D) shows an airway with reduced
lumen, structural reorganization of the airway wall,
increased smooth muscle, and deposition of peri-
bronchial connective tissue.
Printed with permission of Dr. James C. Hogg and Stuart Greene.

peripheral airways, similar to those that occur in the


central airways: exudate of fluid and cells in the
airway wall and lumen, goblet and squamous cell
(A) shows a central bronchus from the lung of a ciga- metaplasia of the epithelium43, edema of the airway
rette smoker with normal lung function. Only small mucosa due to inflammation, and excess mucus in the
amounts of muscle are present and the epithelial airways due to goblet cell metaplasia.
glands are small. This contrasts sharply with a dis-
eased bronchus (B), where the muscle appears as a However, the most characteristic change in the peripheral
thick bundle and the glands are enlarged. (C) shows airways of patients with COPD is airway narrowing.
these enlarged glands at a higher magnification. There Inflammation initiated by cigarette smoking45 and other
is evidence of a chronic inflammatory process involving risk factors75 leads to repeated cycles of injury and
polymorphonuclear (arrowhead) and mononuclear repair of the walls of the peripheral airways. Injury is
cells, including plasma cells (arrow). caused either directly by inhaled toxic particles and
gases such as those found in cigarette smoke, or
Printed with permission of Dr. James C. Hogg and Stuart Greene.
indirectly by the action of inflammatory mediators; this
Peripheral Airways injury then initiates repair processes. Although airway
repair is only partly understood, it seems likely that
The peripheral airways include small bronchi and disordered repair processes can lead to tissue
bronchioles that have an internal diameter of less than remodeling with altered structure and function.
Cigarette smoke may impair lung repair mechanisms,
2 mm (Figure 4-8). The early decline in lung function
thereby further contributing to altered lung structure84-86.
in COPD is correlated with inflammatory changes in the
Even normal lung repair mechanisms can lead to airway

34 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 35

remodeling because tissue repair in the airways, as Figure 4-10. Normal Respiratory Bronchioles
elsewhere in the body, may involve scar tissue formation. and Centrilobular Emphysema
In any case, this injury-and-repair process results in a
structural remodeling of the airway wall, with increasing
collagen content and scar tissue formation, that narrows
the lumen and produces fixed airways obstruction87.
The peripheral airways become the major site of airways
obstruction in COPD, and direct measurements of
peripheral airways resistance88 show that the structural
changes in the airway wall are the most important cause
of the increase in peripheral airways resistance in COPD.
Inflammatory changes such as airway edema and
mucus hypersecretion also contribute to airway narrowing
in COPD. So does loss of elastic recoil, but fibrosis of
the small airways plays the largest role.

Fibrosis in the peripheral airways, as elsewhere in the


body, is characterized by the accumulation of mesenchymal
cells (fibroblasts and myofibroblasts) and extracellular
connective tissue matrix. Several cell types including
mononuclear phagocytes and epithelial cells may produce
mediators that drive this process. The mediators that (A) shows a photomicrograph of the pleural surface
drive the accumulation of these cells and of the matrix of a normal lung, with a secondary lobule defined by
are incompletely defined, but it is likely that several a connective tissue septum (solid arrow) and several
mediators including TGF-ß, ET-1, Insulin-like growth terminal bronchioles (TB) filled with opaque material.
factor-1, fibronectin, platelet-derived growth factor (B) shows a low-power photomicrograph of a normal
(PDGF), and others are involved89. terminal bronchiole (TB) branching into a respiratory
bronchiole (RB), which eventually end in alveolar
Figure 4-9. Normal and Emphysematous Lungs ducts (AD). (C) is a schematic diagram of centrilobu-
lar emphysema and (D) shows the bronchographic
appearance of this lesion (TB=terminal bronchiole;
CLE=centrilobular emphysema).
Printed with permission of Dr. James C. Hogg and Stuart Greene.

common type of parenchymal destruction in COPD


patients is the centrilobular form of emphysema (Figure
4-10), which involves dilatation and destruction of the
respiratory bronchioles90. These lesions occur more
frequently in the upper lung regions in milder cases, but
Photomicrographs of paper-mounted whole lung sec- in advanced disease they may appear diffusely throughout
tions prepared from (A) a normal lung, (B) a lung with the entire lung and also involve destruction of the pulmonary
mild centrilobular emphysema, and (C) a lung with capillary bed. Panacinar emphysema, which extends
severe panacinar emphysema. Note that the centrilob- throughout the acinus, is the characteristic lesion seen in
ular form affects mainly the upper lung regions where- alpha-1 antitrypsin deficiency and involves dilatation and
as the panacinar form is more apparent in the lower destruction of the alveolar ducts and sacs as well as the
lung regions. respiratory bronchioles. It tends to affect the lower more
Printed with permission of Dr. James C. Hogg and Stuart Greene.
than upper lung regions. Because this process usually
affects all of the acini in the secondary lobule, it is also
Lung Parenchyma referred to as panlobular emphysema. The primary
mechanism of lung parenchyma destruction, in both
smoking-related COPD and alpha-1 antitrypsin deficiency,
The lung parenchyma includes the gas exchanging surface
is thought to be an imbalance of endogenous proteinases
of the lung (respiratory bronchioles and alveoli) and the
and antiproteinases in the lung. Oxidative stress, another
pulmonary capillary system (Figure 4-9). The most
consequence of inflammation, may also contribute91.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 35


GOLD_WR_05 8/18/05 12:56 PM Page 36

exercise and then at rest. As COPD worsens, greater


Figure 4-11. Pathological Changes of the Pulmonary
Vasculature in COPD amounts of smooth muscle, proteoglycans, and collagen95
further thicken the vessel wall. In advanced disease, the
changes in the muscular arteries may be associated with
emphysematous destruction of the pulmonary capillary bed.

.
PATHOPHYSIOLOGY
Pathological changes in COPD lead to corresponding
physiological abnormalities that usually become
evident first on exercise and later also at rest.
Physiological changes characteristic of the disease
include mucus hypersecretion, ciliary dysfunction, airflow
limitation, pulmonary hyperinflation, gas exchange
abnormalities, pulmonary hypertension, and cor
pulmonale, and they usually develop in this order
over the course of the disease. In turn, various
physiological abnormalities contribute to the
characteristic symptoms of COPD – chronic cough
and sputum production and dyspnea.

Photomicrographs of small (A) and large (B) vessels Mucus Hypersecretion


in the lung of a heavy smoker with normal lung func- and Ciliary Dysfunction
tion, and small (C) and large (D) vessels in the lung of
a patient with severe emphysema. Note that the small- Mucus hypersecretion in COPD is caused by the
er vessel has thicker walls (compare arrows in A and stimulation of the enlarged mucus secreting glands
C) and that the larger vessel has a thicker media and increased number of goblet cells by inflammatory
(compare arrows in B and D) in the patient with severe mediators such as leukotrienes, proteinases, and
emphysema. (L=vessel lumen; magnification neuropeptides. Ciliated epithelial cells undergo
bars=100 µ). squamous metaplasia leading to impairment in
mucociliary clearance mechanisms. These changes
Printed with permission of Dr. James C. Hogg and Stuart Greene.
are usually the first physiological abnormalities to
develop in COPD, and can be present for many years
Pulmonary Vasculature before any other physiological abnormalities develop.
Pulmonary vascular changes in COPD (Figure 4-11) are
Airflow Limitation and
characterized by a thickening of the vessel wall that
Pulmonary Hyperinflation
begins early in the natural history of the disease, when
lung function is reasonably well maintained and pulmonary
Expiratory airflow limitation is the hallmark physiological
vascular pressures are normal at rest92. Endothelial change of COPD. The airflow limitation characteristic
dysfunction of the pulmonary arteries, which may be of COPD is primarily irreversible, with a small reversible
caused directly by cigarette smoke products93 or indirectly component. Several pathological characteristics
by inflammatory mediators14, occurs early in COPD94. contribute to airflow limitation and changes in pulmonary
Since endothelium plays an important role in regulating mechanics, as summarized in Figure 4-12. The
vascular tone and cell proliferation, it is likely that irreversible component of airflow limitation is primarily
endothelial dysfunction might initiate the sequence of due to remodeling42,43,87,88,96,97 – fibrosis and narrowing – of
events that results ultimately in structural changes. the small airways that produces fixed airways obstruction
Thickening of the intima is the first structural change92, and a consequent increase in airways resistance. The
followed by an increase in vascular smooth muscle and sites of airflow limitation in COPD are the smaller
the infiltration of the vessel wall by inflammatory cells, conducting airways, including bronchi and bronchioles
including macrophages and CD8+ T lymphocytes14. less than 2 mm in internal diameter. In the normal lung,
These structural changes are correlated with an increase resistance of these smaller airways makes up a small
in pulmonary vascular pressure that develops first with percentage of the total airways resistance88. But in

36 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 37

Figure 4-12. Causes of Airflow Limitation in COPD forced expiratory volume in one second (FEV1) and
forced vital capacity (FVC). As COPD progresses, with
increased thickness of the airway wall, loss of alveolar
Irreversible • Fibrosis and narrowing of airways attachments, and loss of lung elastic recoil, FEV1 and
• Loss of elastic recoil due to alveolar FVC decrease. A decrease in the ratio of FEV1 to FVC
destruction is often the first sign of developing airflow limitation.
• Destruction of alveolar support that FEV1 declines naturally with age, but the rate of decline
maintains patency of small airways in COPD patients is generally greater than that in normal
subjects.
Reversible • Accumulation of inflammatory cells,
mucus, and plasma exudate in bronchi With increasing severity of airflow limitation, expiration
• Smooth muscle contraction in peripheral becomes flow-limited during tidal breathing. Initially, this
and central airways occurs only during exercise, but later it is also seen at
• Dynamic hyperinflation during exercise rest. In parallel with this, functional residual capacity (FRC)
increases due to the combination of the decrease in the
elastic properties of the lungs, premature airway closure,
patients with COPD the total lower airways resistance and a variable dynamic element reflecting the breathing
approximately doubles, and most of the increase is due
pattern adopted to cope with impaired lung mechanics.
to a large increase in peripheral airways resistance88.
As airflow limitation develops, the rate of lung emptying is
Although some have argued that a larger proportion of
slowed and the interval between inspiratory efforts does
the total resistance should be attributed to peripheral air-
not allow expiration to the relaxation volume of the
ways in the normal lung, there is wide agreement that the
respiratory system; this leads to dynamic pulmonary
peripheral airways become the major site of obstruction
hyperinflation. The increase in FRC can impair inspiratory
in COPD.
muscle function and coordination, although the contractility
of the diaphragm, when normalized for lung volume,
Parenchymal destruction (emphysema) plays a smaller seems to be preserved. These changes occur as the
role in this irreversible component but contributes to disease advances but are almost always seen first during
expiratory airflow limitation and the increase in airways exercise, when the greater metabolic stimulus to ventilation
resistance in several ways. Destruction of alveolar stresses the ability of the ventilatory pump to maintain
attachments inhibits the ability of the small airways to gas exchange.
maintain patency98. Alveolar destruction is also associated
with a loss of elastic recoil of the lung99,100, which decreases Gas Exchange Abnormalities
the intra-alveolar pressure driving exhalation.
In advanced COPD, peripheral airways obstruction,
Although both the destruction of alveolar attachments to parenchymal destruction, and pulmonary vascular
the outer wall of the peripheral airways and the loss of abnormalities reduce the lung's capacity for gas
lung elastic recoil produced by emphysema have been exchange, producing hypoxemia and, later on, hypercapnia.
implicated in the pathogenesis of peripheral airways The correlation between routine lung function tests and
obstruction98,100, direct measurements of peripheral air- arterial blood gases is poor, but significant hypoxemia or
ways resistance88 show that the structural changes in the hypercapnia is rare when FEV1 is greater than 1.00 L102.
airway wall are the most important cause of the increase Hypoxemia is initially only present during exercise, but as
in peripheral airways resistance in COPD. the disease continues to progress it is also present at rest.

Airway smooth muscle contraction, ongoing airway Inequality in the ventilation/perfusion ratio (VA/Q) is the
inflammation, and intraluminal accumulation of mucus major mechanism behind hypoxemia in COPD, regardless
and plasma exudate may be responsible for the small of the stage of the disease103. In the peripheral airways,
part of airflow limitation that is reversible with treatment. injury of the airway wall is associated with VA /Q
Inflammation and accumulation of mucus and exudate mismatching, as indicated by a significant correlation
may be particularly important during exacerbations101. between bronchiolar inflammation and the distribution of
ventilation. In the parenchyma, destruction of the lung
Airflow limitation in COPD is best measured through surface area by emphysema reduces diffusing capacity
spirometry, which is key to the diagnosis and management and interferes with gas exchange104. High VA/Q units
of the disease. The essential spirometric measurements probably represent emphysematous regions with alveolar
for diagnosis and monitoring of COPD patients are the destruction and loss of pulmonary vasculature. The

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 37


GOLD_WR_05 8/18/05 12:56 PM Page 38

severity of pulmonary emphysema appears to be related promoting remodeling of the vessel wall (either by inducing
to the overall inefficiency of the lung as a gas exchanger. the release of growth factors107 or as a consequence of
This is reflected by the good correlation between the the mechanical stress that results from hypoxic
diffusing capacity of carbon monoxide per liter of alveolar vasoconstriction).
volume (DLco/VA) and the severity of macroscopic
emphysema. Reduced ventilation due to loss of elastic Pulmonary hypertension is associated with the development
recoil in the emphysematous lung, together with the loss of cor pulmonale, defined as "hypertrophy of the right
of the capillary bed and the generalized inhomogeneity ventricle resulting from diseases affecting the function
of ventilation due to the patchy nature of these changes, and/or structure of the lungs, except when these pulmonary
leads to areas of VA/Q mismatching that result in arterial alterations are the result of diseases that primarily affect
hypoxemia. the left side of the heart, as in congenital heart disease."
This is a pathological definition and the clinical diagnosis
The relationship between pulmonary vascular abnormalities and assessment of right ventricular hypertrophy is difficult
and VA/Q relationships has been investigated in patients in life.
with mild COPD. The more severe the vessel wall damage
is, the less the reversal of hypoxic vasoconstriction by The prevalence and natural history of cor pulmonale in
oxygen105. This suggests that pathology in the pulmonary COPD are not yet clear. Pulmonary hypertension and
artery wall, particularly when it affects the intimal layer, reduction of the vascular bed due to emphysema can
may play a key role in determining the loss of vascular lead to right ventricular hypertrophy and right heart failure,
response to hypoxia that contributes to VA/Q mismatching. but right ventricular function appears to be maintained
Chronic hypercapnia usually reflects inspiratory muscle in some patients despite the presence of pulmonary
dysfunction and alveolar hypoventilation. hypertension108. Right heart failure is associated with
venous stasis and thrombosis that may result in pulmonary
Pulmonary Hypertension and Cor Pulmonale embolism and further compromise the pulmonary
circulation.
Pulmonary hypertension develops late in the course of
COPD (Stage IV: Very Severe COPD), usually after the Systemic Effects
development of severe hypoxemia (PaO2 < 8.0 kPa or 60
mm Hg) and often hypercapnia as well. It is the major COPD is associated with systemic (i.e., extrapulmonary)
cardiovascular complication of COPD and is associated effects, such as systemic inflammation and skeletal
with the development of cor pulmonale and with a poor muscle dysfunction. Evidence of systemic inflammation
prognosis106. However, even in patients with severe includes the presence of systemic oxidative stress109,
disease, pulmonary arterial pressure is usually only abnormal concentrations of circulating cytokines110, and
modestly elevated at rest, though it may rise markedly activation of inflammatory cells111,112. Evidence of skeletal
with exercise. Pulmonary hypertension in COPD is muscle dysfunction includes the progressive loss of
believed to progress rather slowly even if left untreated. skeletal muscle mass and the presence of several
Further studies are required to firmly establish the natural bioenergetic abnormalities113. These systemic effects have
history of pulmonary hypertension in COPD. important clinical consequences, as they contribute to the
limitation of patients' exercise capacity and thus the
Factors that are known to contribute to the development decline of health status in COPD. The presence of these
of pulmonary hypertension in patients with COPD include systemic effects appears to worsen a patient's prognosis114.
vasoconstriction; remodeling of pulmonary arteries,
which thickens the vessel walls and reduces the lumen; Pathophysiology and the
and destruction of the pulmonary capillary bed by Symptoms of COPD
emphysema, which further increases the pressure
required to perfuse the pulmonary vascular bed. Chronic cough and sputum production, sometimes labeled
Vasoconstriction may itself have several causes, including as chronic bronchitis, are a result of airway inflammation,
hypoxia, which causes pulmonary vascular smooth muscle which leads to mucus hypersecretion and dysfunction of
to contract; impaired mechanisms of endothelium-dependent the normal ciliary clearance mechanisms. Sputum is
vasodilation, such as reduced NO synthesis or release; produced in COPD as a result of the inflammatory
and abnormal secretion of vasoconstrictor peptides (such response, and contains plasma proteins exuded from the
as ET-1, which is produced by inflammatory cells). In microvessels of the bronchial circulation, inflammatory
advanced COPD, hypoxia plays the primary role in cells, and small amounts of mucus from epithelial goblet
producing pulmonary hypertension, both by causing cells. The volume of sputum produced overpowers clearance
vasoconstriction of the pulmonary arteries and by mechanisms, resulting in cough and expectoration.

38 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 39

Some pathological abnormalities, such as inflammation of greater oxygen consumption, decreased mixed venous
the submucosal glands and hyperplasia of goblet cells, oxygen tension, and further amplification of gas
may contribute to chronic sputum production, although exchange abnormalities120. Worsening of VA/Q relation-
these pathological abnormalities are not present in all ships has several causes in exacerbations. Airway
patients with this symptom. inflammation and edema, mucus hypersecretion, and
bronchoconstriction may contribute to changes in the
Dyspnea, an abnormal awareness of the act of breathing, distribution of ventilation, while hypoxic constriction of
usually reflects an imbalance between the neural drive to pulmonary arterioles may modify the distribution of
the respiratory muscles and the effectiveness of the perfusion. Additional contributors to worsening gas
resulting ventilation. Different individuals use different exchange in exacerbations include abnormal patterns of
words to describe the feeling of breathlessness, which is breathing and fatigue of the respiratory muscles. These
also influenced by other factors such as mood. In COPD can cause further deterioration in blood gases and
patients, dyspnea is mainly the result of impaired lung worsening of respiratory acidosis, leading to severe
mechanics (increased airways resistance, decreased respiratory failure and death120-123. Alveolar hypoventilation
elastic recoil). It is only present on vigorous exercise in also contributes to hypoxemia, hypercapnia, and
the early stages of disease but may be present at rest as respiratory acidosis. In turn, hypoxemia and respiratory
the mechanical impairment becomes severe. acidosis promote pulmonary vasoconstriction, which
increases pulmonary artery pressures and imposes an
added load on the right ventricle.
PATHOLOGY AND
REFERENCES
PATHOPHYSIOLOGY OF
EXACERBATIONS 1. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar
inflammation and its relation to emphysema in smokers.
The progressive course of COPD is complicated by Am J Respir Crit Care Med 1995; 152:1666-72.
exacerbations that have many causes and occur with 2. O'Shaughnessy TC, Ansari TW, Barnes NC, Jeffery PK.
increasing frequency as the disease progresses. Inflammation in bronchial biopsies of subjects with chronic
bronchitis: inverse relationship of CD8+ T lymphocytes with
Pathology FEV1. Am J Respir Crit Care Med 1997; 155:852-7.
3. Di Stefano A, Capelli A, Lusuardi M, Balbo P, Vecchio C,
Distinguishing the pathology of these acute events from Maestrelli P, et al. Severity of airflow limitation is associated
that of the underlying disease is difficult because patients with severity of airway inflammation in smokers. Am J
experiencing an exacerbation are usually too ill to study. Respir Crit Care Med 1998; 158:1277-85.
The limited evidence available suggests that mild COPD
4. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences
exacerbations are associated with increases of both
in interleukin-8 and tumor necrosis factor-alpha in induced
neutrophils and eosinophils in sputum and biopsies, while
sputum from patients with chronic obstructive pulmonary
severe COPD exacerbations are associated with an
disease or asthma. Am J Respir Crit Care Med 1996;
increase in sputum neutrophils and eosinophils18,19. At
153:530-4.
least in sputum, the changes in inflammatory cells during
5. Thompson AB, Daughton D, Robbins RA, Ghafouri MA,
exacerbations of COPD are the same as those observed
Oehlerking M, Rennard SI. Intraluminal airway inflamma-
during exacerbations of asthma115-119. So far no study has
tion in chronic bronchitis. Characterization and correlation
been conducted examining the pathological abnormalities
with clinical parameters. Am Rev Respir Dis 1989;
associated with fatal exacerbations of COPD, which can be
140:1527-37.
considered the extreme end of the spectrum of severity.
6. Lacoste JY, Bousquet J, Chanez P, Van Vyve T, Simony-
Pathophysiology Lafontaine J, Lequeu N, et al. Eosinophilic and neutrophilic
inflammation in asthma, chronic bronchitis, and chronic
Expiratory airflow is almost unchanged during mild obstructive pulmonary disease. J Allergy Clin Immunol
exacerbations18, and only slightly reduced during severe 1993; 92:537-48.
exacerbations120,121. Although the pathophysiology of 7. Beeh KM, Beier J, Kommann O, Mander A, Buhl R. Long-
exacerbations is not fully understood, the primary term repeatability of induced sputum cells and inflammato-
physiological change in severe exacerbations is a further ry markers in stable, moderately severe COPD. Chest
worsening of gas exchange, primarily produced by 2003; 123:778-83.
increased VA/Q inequality. As VA/Q relationships worsen, 8. Stockley RA. Neutrophils and the pathogenesis of COPD.
increased work of the respiratory muscles results in Chest 2002; 121:151S-155S.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 39


GOLD_WR_05 8/18/05 12:56 PM Page 40

9. Peleman RA, Rytila PH, Kips JC, Joos GF, Pauwels RA. 23. Shapiro SD. Matrix metalloproteinase degradation of extra-
The cellular composition of induced sputum in chronic cellular matrix: biological consequences. Curr Opin Cell
obstructive pulmonary disease. Eur Respir J 1999; 13:839-43. Biol 1998; 10:602-8.
10. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, 24. Warren JS, Ward PA. Consequences of oxidant injury. In:
Mapp CE, et al. CD8+ T-lymphocytes in peripheral airways Crystal RG, West JB, eds. The lung: scientific foundations.
of smokers with chronic obstructive pulmonary disease. Philadelphia: Lippincott-Raven; 1997. p. 2279-88.
Am J Respir Crit Care Med 1998; 157:822-6. 25. Ganz T, Lehrer RI. Defensins. Curr Opin Immunol 1994;
11. Roth MD, Arora A, Barsky SH, Kleerup EC, Simmons M, 6:584-9.
Tashkin DP. Airway inflammation in young marijuana and 26. Hill AT, Bayley D, Stockley RA. The interrelationship of spu-
tobacco smokers. Am J Respir Crit Care Med 1998; tum inflammatory markers in patients with chronic bronchi-
157:928-37. tis. Am J Respir Crit Care Med 1999; 160:893-8.
12. Keatings VM, Barnes PJ. Granulocyte activation markers in 27. Yamamoto C, Yoneda T, Yoshikawa M, Fu A, Tokuyama T,
induced sputum: comparison between chronic obstructive Tsukaguchi K, et al. Airway inflammation in COPD
pulmonary disease, asthma, and normal subjects. Am J assessed by sputum levels of interleukin-8. Chest 1997;
Respir Crit Care Med 1997; 155:449-53. 112:505-10.
13. Balbi B, Bason C, Balleari E, Fiasella F, Pesci A, Ghio R, 28. Senior RM, Griffin GL, Mecham RP. Chemotactic activity of
et al. Increased bronchoalveolar granulocytes and granulo- elastin-derived peptides. J Clin Invest 1980; 66:859-62.
cyte/macrophage colony-stimulating factor during exacer- 29. Heise CE, O'Dowd BF, Figueroa DJ, Sawyer N, Nguyen T,
bations of chronic bronchitis. Eur Respir J 1997; 10:846-50. Im DS, et al. Characterization of the human cysteinyl
14. Peinado VI, Barbera JA, Abate P, Ramirez J, Roca J, leukotriene 2 receptor. J Biol Chem 2000; 275:30531-6.
Santos S, et al. Inflammatory reaction in pulmonary muscular 30. de Godoy I, Donahoe M, Calhoun WJ, Mancino J, Rogers
arteries of patients with mild chronic obstructive pulmonary RM. Elevated TNF-alpha production by peripheral blood
disease. Am J Respir Crit Care Med 1999; 159:1605-11. monocytes of weight-losing COPD patients. Am J Respir
15. Liu AN, Mohammed AZ, Rice WR, Fiedeldey DT, Crit Care Med 1996; 153:633-7.
Liebermann JS, Whitsett JA, et al. Perforin-independent 31. Capelli A, Di Stefano A, Gnemmi I, Balbo P, Cerutti CG,
CD8(+) T-cell-mediated cytotoxicity of alveolar epithelial Balbi B, et al. Increased MCP-1 and MIP-1beta in bron-
cells is preferentially mediated by tumor necrosis factor- choalveolar lavage fluid of chronic bronchitics. Eur Respir J
alpha: relative insensitivity to Fas ligand. Am J Respir Cell 1999; 14:160-5.
Mol Biol 1999; 20:849-58. 32. Hoshi H, Ohno I, Honma M, Tanno Y, Yamauchi K, Tamura
16. Mueller R, Chanez P, Campbell AM, Bousquet J, Heusser G, et al. IL-5, IL-8 and GM-CSF immunostaining of sputum
C, Bullock GR. Different cytokine patterns in bronchial cells in bronchial asthma and chronic bronchitis. Clin Exp
biopsies in asthma and chronic bronchitis. Respir Med Allergy 1995; 25:720-8.
1996; 90:79-85. 33. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace
17. Liu H, Lazarus SC, Caughey GH, Fahy JV. Neutrophil elas- E, Rizzo A, et al. Transforming growth factor-beta expres-
tase and elastase-rich cystic fibrosis sputum degranulate sion in mucosal biopsies in asthma and chronic bronchitis.
human eosinophils in vitro. Am J Respir Crit Care Med 1997; 156:591-9.
Am J Physiol 1999; 276:L28-34. 34. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk
18. Saetta M, Di Stefano A, Maestrelli P, Turato G, Ruggieri J, Hiemstra PS, et al. Transforming growth factor beta1 and
MP, Roggeri A, et al. Airway eosinophilia in chronic bron- recruitment of macrophages and mast cells in airways in
chitis during exacerbations. Am J Respir Crit Care Med chronic obstructive pulmonary disease. Am J Respir Crit
1994; 150:1646-52. Care Med 1998; 158:1951-7.
19. Saetta M, Di Stefano A, Maestrelli P, Turato G, Mapp CE, 35. Chalmers GW, Macleod KJ, Sriram S, Thomson LJ,
Pieno M, et al. Airway eosinophilia and expression of inter- McSharry C, Stack BH, et al. Sputum endothelin-1 is
leukin-5 protein in asthma and in exacerbations of chronic increased in cystic fibrosis and chronic obstructive pul-
bronchitis. Clin Exp Allergy 1996; 26:766-74. monary disease. Eur Respir J 1999; 13:1288-92.
20. Mills PR, Davies RJ, Devalia JL. Airway epithelial cells, 36. Fujii T, Otsuka T, Tanaka S, Kanazawa H, Hirata K, Kohno
cytokines, and pollutants. Am J Respir Crit Care Med M, et al. Plasma endothelin-1 level in chronic obstructive
1999; 160:S38-43. pulmonary disease: relationship with natriuretic peptide.
21. Di Stefano A, Maestrelli P, Roggeri A, Turato G, Calabro S, Respiration 1999; 66:212-9.
Potena A, et al. Up-regulation of adhesion molecules in the 37. Tomaki M, Ichinose M, Miura M, Hirayama Y, Yamauchi H,
bronchial mucosa of subjects with chronic obstructive bron- Nakajima N, et al. Elevated substance P content in
chitis. Am J Respir Crit Care Med 1994; 149:803-10. induced sputum from patients with asthma and patients
22. McElvaney NG, Crystal RG. Proteinases and lung injury. with chronic bronchitis. Am J Respir Crit Care Med 1995;
In: Crystal RG, West JB, eds. The lung: scientific founda- 151:613-7.
tions. Philadelphia: Lippincott-Raven; 1997. p. 2205-18.

40 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 41

38. Lucchini RE, Facchini F, Turato G, Saetta M, Caramori G, 54. Masubuchi T, Koyama S, Sato E, Takamizawa A, Kubo K,
Ciaccia A, et al. Increased VIP-positive nerve fibers in the Sekiguchi M, et al. Smoke extract stimulates lung epithelial
mucous glands of subjects with chronic bronchitis. Am J cells to release neutrophil and monocyte chemotactic
Respir Crit Care Med 1997; 156:1963-8. activity. Am J Pathol 1998; 153:1903-12.
39. Chanez P, Springall D, Vignola AM, Moradoghi-Hattvani A, 55. Speizer FE, Tager IB. Epidemiology of chronic mucus
Polak JM, Godard P, et al. Bronchial mucosal immunoreac- hypersecretion and obstructive airways disease. Epidemiol
tivity of sensory neuropeptides in severe airway diseases. Rev 1979; 1:124-42.
Am J Respir Crit Care Med 1998; 158:985-90. 56. Turato G, Di Stefano A, Maestrelli P, Mapp CE, Ruggieri
40. Metcalf JP, Thompson AB, Gossman GL, Nelson KJ, MP, Roggeri A, et al. Effect of smoking cessation on airway
Koyama S, Rennard SI, et al. Gcglobulin functions as a inflammation in chronic bronchitis. Am J Respir Crit Care
cochemotaxin in the lower respiratory tract. A potential Med 1995; 152:1262-7.
mechanism for lung neutrophil recruitment in cigarette 57. Li XY, Brown D, Smith S, MacNee W, Donaldson K. Short-
smokers. Am Rev Respir Dis 1991; 143:844-9. term inflammatory responses following intratracheal instilla-
41. Wright JL, Lawson LM, Pare PD, Wiggs BJ, Kennedy S, tion of fine and ultrafine carbon black in rats Inhal Toxicol
Hogg JC. Morphology of peripheral airways in current 1999; 11:709-31.
smokers and ex-smokers. Am Rev Respir Dis 1983; 58. Monn C, Becker S. Cytotoxicity and induction of proinflam-
127:474-7. matory cytokines from human monocytes exposed to fine
42. Mullen JB, Wright JL, Wiggs BR, Pare PD, Hogg JC. (PM2.5) and coarse particles (PM10-2.5) in outdoor and
Reassessment of inflammation of airways in chronic bron- indoor air. Toxicol Appl Pharmacol 1999; 155:245-52.
chitis. BMJ (Clin Res Ed) 1985; 291:1235-9. 59. Salvi S, Blomberg A, Rudell B, Kelly F, Sandstrom T,
43. Cosio M, Ghezzo H, Hogg JC, Corbin R, Loveland M, Holgate ST, et al. Acute inflammatory responses in the air-
Dosman J, et al. The relations between structural changes ways and peripheral blood after short-term exposure to
in small airways and pulmonary-function tests. N Engl J diesel exhaust in healthy human volunteers. Am J Respir
Med 1978; 298:1277-81. Crit Care Med 1999; 159:702-9.
44. McLean KA. Pathogenesis of pulmonary emphysema. Am 60. Von Essen SG, O'Neill DP, McGranaghan S, Olenchock
J Med 1958; 25:62-74. SA, Rennard SI. Neutrophilic respiratory tract inflammation
45. Niewoehner DE, Kleinerman J, Rice DB. Pathologic and peripheral blood neutrophilia after grain sorghum dust
changes in the peripheral airways of young cigarette smok- extract challenge. Chest 1995; 108:1425-33.
ers. N Engl J Med 1974; 291:755-8. 61. Von Essen SG, Robbins RA, Thompson AB, Ertl RF, Linder
46. Auerbach O, Garfinkel L, Hammond EC. Relation of smok- J, Rennard SI. Mechanisms of neutrophil recruitment to the
ing and age to findings in lung parenchyma: a microscopic lung by grain dust exposure [published erratum appears in
study. Chest 1974; 65:29-35. Am Rev Respir Dis 1989; 139:1065]. Am Rev Respir Dis
47. Auerbach O, Hammond EC, Garfinkel L, Benante C. 1988; 138:921-7.
Relation of smoking and age to emphysema. Whole-lung 62. Laurell CB, Eriksson S. The electrophoretic alpha-1 globu-
section study. N Engl J Med 1972; 286:853-7. lin pattern of serum in alpha-1 antitrypsin deficiency. Scand
48. Petty TL, Silvers GW, Stanford RE, Baird MD, Mitchell RS. J Clin Lab Invest 1963; 15:132-40.
Small airway pathology is related to increased closing 63. Carp H, Miller F, Hoidal JR, Janoff A. Potential mechanism
capacity and abnormal slope of phase III in excised human of emphysema: alpha 1-proteinase inhibitor recovered from
lungs. Am Rev Respir Dis 1980; 121:449-56. lungs of cigarette smokers contains oxidized methionine
49. Ollerenshaw SL, Woolcock AJ. Characteristics of the and has decreased elastase inhibitory capacity. Proc Natl
inflammation in biopsies from large airways of subjects Acad Sci U S A 1982; 79:2041-5.
with asthma and subjects with chronic airflow limitation. 64. Cohen AB, James HL. Reduction of the elastase inhibitory
Am Rev Respir Dis 1992; 145:922-7. capacity of alpha 1-antitrypsin by peroxides in cigarette
50. Gadek JE, Fells GA, Crystal RG. Cigarette smoking smoke: an analysis of brands and filters. Am Rev Respir
induces functional antiproteinase deficiency in the lower Dis 1982; 126:25-30.
respiratory tract of humans. Science 1979; 206:1315-6. 65. Shapiro SD. Elastolytic metalloproteinases produced by
51. Stone PJ, Calore JD, McGowan SE, Bernardo J, Snider human mononuclear phagocytes. Potential roles in
GL, Franzblau C. Functional alpha 1-proteinase inhibitor in destructive lung disease. Am J Respir Crit Care Med 1994;
the lower respiratory tract of cigarette smokers is not 150:S160-4.
decreased. Science 1983; 221:1187-9. 66. Sommerhoff CP, Nadel JA, Basbaum CB, Caughey GH.
52. Hunninghake GW, Crystal RG. Cigarette smoking and lung Neutrophil elastase and cathepsin G stimulate secretion
destruction. Accumulation of neutrophils in the lungs of cig- from cultured bovine airway gland serous cells. J Clin
arette smokers. Am Rev Respir Dis 1983; 128:833-8. Invest 1990; 85:682-9.
53. Mio T, Romberger DJ, Thompson AB, Robbins RA, Heires 67. Witko-Sarsat V, Halbwachs-Mecarelli L, Schuster A,
A, Rennard SI. Cigarette smoke induces interleukin-8 Nusbaum P, Ueki I, Canteloup S, et al. Proteinase 3, a
release from human bronchial epithelial cells. Am J Respir potent secretagogue in airways, is present in cystic fibrosis
Crit Care Med 1997; 155:1770-6. sputum. Am J Respir Cell Mol Biol 1999; 20:729-36.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 41


GOLD_WR_05 8/18/05 12:56 PM Page 42

68. Christensen TG, Korthy AL, Snider GL, Hayes JA. 83. Rogers DF, Jeffery PK. Inhibition by oral N-acetylcysteine
Irreversible bronchial goblet cell metaplasia in hamsters of cigarette smoke-induced "bronchitis" in the rat. Exp Lung
with elastase-induced panacinar emphysema. J Clin Invest Res 1986; 10:267-83.
1977; 59:397-404. 84. Nakamura Y, Romberger DJ, Tate L, Ertl RF, Kawamoto M,
69. Dekhuijzen PN, Aben KK, Dekker I, Aarts LP, Wielders PL, Adachi Y, et al. Cigarette smoke inhibits lung fibroblast pro-
van Herwaarden CL, et al. Increased exhalation of hydro- liferation and chemotaxis. Am J Respir Crit Care Med
gen peroxide in patients with stable and unstable chronic 1995; 151:1497-503.
obstructive pulmonary disease. Am J Respir Crit Care Med 85. Osman M, Cantor JO, Roffman S, Keller S, Turino GM,
1996; 154:813-6. Mandl I. Cigarette smoke impairs elastin resynthesis in
70. Maziak W, Loukides S, Culpitt S, Sullivan P, Kharitonov SA, lungs of hamsters with elastase-induced emphysema. Am
Barnes PJ. Exhaled nitric oxide in chronic obstructive pul- Rev Respir Dis 1985; 132:640-3.
monary disease. Am J Respir Crit Care Med 1998; 86. Laurent P, Janoff A, Kagan HM. Cigarette smoke blocks
157:998-1002. cross-linking of elastin in vitro. Chest 1983; 83:63-5S.
71. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi 87. Matsuba K, Thurlbeck WM. The number and dimensions of
M, Kharitonov SA, et al. Exhaled 8-isoprostane as an in small airways in emphysematous lungs. Am J Pathol 1972;
vivo biomarker of lung oxidative stress in patients with 67:265-75.
COPD and healthy smokers. Am J Respir Crit Care Med 88. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of
2000; 162:1175-7. airway obstruction in chronic obstructive lung disease. N
72. Pratico D, Basili S, Vieri M, Cordova C, Violi F, Fitzgerald Engl J Med 1968; 278:1355-60.
GA. Chronic obstructive pulmonary disease is associated 89. Rennard SI. Inflammation and repair processes in chronic
with an increase in urinary levels of isoprostane F2alpha- obstructive pulmonary disease. Am J Respir Crit Care Med
III, an index of oxidant stress. Am J Respir Crit Care Med 1999; 160:S12-6.
1998; 158:1709-14. 90. Leopold JG, Goeff J. Centrilobular form of hypertrophic
73. Kawikova I, Barnes PJ, Takahashi T, Tadjkarimi S, Yacoub emphysema and its relation to chronic bronchitis. Thorax
MH, Belvisi MG. 8-Epi-PGF2 alpha, a novel noncyclooxy- 1957; 12:219-35.
genase-derived prostaglandin, constricts airways in vitro. 91. Repine JE, Bast A, Lankhorst I. Oxidative stress in chronic
Am J Respir Crit Care Med 1996; 153:590-6. obstructive pulmonary disease. Oxidative Stress Study
74. Ciba Guest Symposium Report. Terminiology, definitions Group. Am J Respir Crit Care Med 1997; 156:341-57.
and classifications of chronic pulmonary emphysema and 92. Wright JL, Lawson L, Pare PD, Hooper RO, Peretz DI,
related conditions. Thorax 1959; 14:286-99. Nelems JM, et al. The structure and function of the pul-
75. Pride NB, Burrows B. Development of impaired lung func- monary vasculature in mild chronic obstructive pulmonary
tion: natural history and risk factors. In: Calverley PM, disease. The effect of oxygen and exercise. Am Rev Respir
Pride NB, eds. Chronic obstructive lung disease. London Dis 1983; 128:702-7.
and Glasgow: Chapman and Hall Medical; 1995. p. 69-71. 93. Sekhon HS, Wright JL, Churg A. Cigarette smoke causes
76. Saetta M, Di Stefano A, Maestrelli P, Ferraresso A, Drigo rapid cell proliferation in small airways and associated pul-
R, Potena A, et al. Activated T-lymphocytes and monary arteries. Am J Physiol 1994; 267:L557-63.
macrophages in bronchial mucosa of subjects with chronic 94. Peinado VI, Barbera JA, Ramirez J, Gomez FP, Roca J,
bronchitis. Am Rev Respir Dis 1993; 147:301-6. Jover L, et al. Endothelial dysfunction in pulmonary arteries
77. Reid L. Measurement of the bronchial mucous gland layer: of patients with mild COPD. Am J Physiol 1998; 274:L908-13.
a diagnostic yardstick in chronic bronchitis. Thorax 1960; 95. Riley DJ, Thakker-Varia S, Poiani GJ, Tozzi CA. Vascular
15:132-41. remodeling. In: Crystal RG, West JB, Barnes PJ, Weibel
78. Jamal K, Cooney TP, Fleetham JA, Thurlbeck WM. Chronic ER, eds. The lung: scientific foundations. Philadelphia:
bronchitis. Correlation of morphologic findings to sputum Lippincott-Raven; 1977. p. 1589-97.
production and flow rates. Am Rev Respir Dis 1984; 96. Kuwano K, Bosken CH, Pare PD, Bai TR, Wiggs BR, Hogg
129:719-22. JC. Small airways dimensions in asthma and in chronic
79. Haraguchi M, Shimura S, Shirato K. Morphometric analysis
obstructive pulmonary disease. Am Rev Respir Dis 1993;
of bronchial cartilage in chronic obstructive pulmonary dis-
148:1220-5.
ease and bronchial asthma. Am J Respir Crit Care Med
97. Matsuba K, Wright JL, Wiggs BR, Pare PD, Hogg JC. The
1999; 159:1005-13.
changes in airways structure associated with reduced
80. Thurlbeck WM, Pun R, Toth J, Frazer RG. Bronchial carti-
forced expiratory volume in one second. Eur Respir J
lage in chronic obstructive lung disease. Am Rev Respir
1989; 2:834-9.
Dis 1974; 109:73-80.
98. Dayman H. Mechanics of airflow in health and emphyse-
81. Snider GL. Parker B. Francis Lecture. Animal models of
ma. J Clin Invest 1951; 30:1175-90.
chronic airways injury. Chest 1992; 101:74-9S.
82. Snider GL, Faling LJ, Rennard SI. Chronic bronchitis and 99. Butler J, Caro C, Alkaler R, Dubois AB. Physiological fac-
emphysema. In: Murray JF, Nadel JA, eds. Textbook of res- tors affecting airway resistance in normal subjects and in
piratory medicine. Philadelphia: WB Saunders; 2000. p. patients with obstructive airways disease. J Clin Invest
1187-246. 1960; 39:584-91.

42 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 43

100. Mead J, Turner JM, Macklem PT, Little JB. Significance of 114. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss
the relationship between lung recoil and maximum expira- is a reversible factor in the prognosis of chronic obstructive
tory flow. J Appl Physiol 1967; 22:95-108. pulmonary disease. Am J Respir Crit Care Med 1998;
101. Burnett D, Stockley RA. Serum and sputum alpha 2 157:1791-7.
macroglobulin in patients with chronic obstructive airways 115. Pizzichini MM, Pizzichini E, Efthimiadis A, Clelland L,
disease. Thorax 1981; 36:512-6. Mahony JB, Dolovich J, et al. Markers of inflammation in
102. Lane DJ, Howell JB, Giblin B. Relation between airways induced sputum in acute bronchitis caused by Chlamydia
obstruction and CO2 tension in chronic obstructive airways pneumoniae. Thorax 1997; 52:929-31; discussion 6-7.
disease. BMJ 1968; 3:707-9. 116. Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K,
103. Rodriguez-Roisin R, MacNee W. Pathophysiology of chron- Gleich GJ, Berman L, et al. Sputum eosinophilia predicts
ic obstructive pulmonary disease. In: Postma DS, Siafakas benefit from prednisone in smokers with chronic obstruc-
NM, eds. Management of chronic obstructive pulmonary tive bronchitis. Am J Respir Crit Care Med 1998; 158:1511-7.
disease. European Respiratory Monograph 1998; 3:107-26. 117. Maestrelli P, Saetta M, Di Stefano A, Calcagni PG, Turato
104. McLean A, Warren PM, Gillooly M, MacNee W, Lamb D. G, Ruggieri MP, et al. Comparison of leukocyte counts in
Microscopic and macroscopic measurements of emphyse- sputum, bronchial biopsies, and bronchoalveolar lavage.
ma: relation to carbon monoxide gas transfer. Thorax 1992; Am J Respir Crit Care Med 1995; 152:1926-31.
47:144-9. 118. Turner MO, Hussack P, Sears MR, Dolovich J, Hargreave
105. Barbera JA, Riverola A, Roca J, Ramirez J, Wagner PD, FE. Exacerbations of asthma without sputum eosinophilia.
Ros D, et al. Pulmonary vascular abnormalities and ventila- Thorax 1995; 50:1057-61.
tion-perfusion relationships in mild chronic obstructive pul- 119. Fahy JV, Kim KW, Liu J, Boushey HA. Prominent neu-
monary disease. Am J Respir Crit Care Med 1994; trophilic inflammation in sputum from subjects with asthma
149:423-9. exacerbation. J Allergy Clin Immunol 1995; 95:843-52.
106. MacNee W. Pathophysiology of cor pulmonale in chronic 120. Barbera JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N,
obstructive pulmonary disease. Part two. Am J Respir Crit et al. Mechanisms of worsening gas exchange during
Care Med 1994; 150:1158-68. acute exacerbations of chronic obstructive pulmonary dis-
107. Knighton DR, Hunt TK, Scheuenstuhl H, Halliday BJ, Werb ease. Eur Respir J 1997; 10:1285-91.
Z, Banda MJ. Oxygen tension regulates the expression of 121. Seemungal TA, Donaldson GC, Bhowmik A, Jeffries DJ,
angiogenesis factor by macrophages. Science 1983; Wedzicha JA. Time course and recovery of exacerbations
221:1283-5. in patients with chronic obstructive pulmonary disease. Am
108. Biernacki W, Flenley DC, Muir AL, MacNee W. Pulmonary J Respir Crit Care Med 2000; 161:1608-13.
hypertension and right ventricular function in patients with 122. Schmidt GA, Hall JB. Acute or chronic respiratory failure.
COPD. Chest 1988; 94:1169-75. Assessment and management of patients with COPD in
109. Rahman I, Morrison D, Donaldson K, MacNee W. Systemic the emergency setting. JAMA 1989; 261:3444-53.
oxidative stress in asthma, COPD, and smokers. Am J 123. Rodriguez-Roisin R. Pulmonary gas exchange in acute res-
Respir Crit Care Med 1996; 154:1055-60. piratory failure. Eur J Anaesthesiol 1994; 11:5-13.
110. Schols AM, Buurman WA, Staal van den Brekel AJ,
Dentener MA, Wouters EF. Evidence for a relation between
metabolic derangements and increased levels of inflamma-
tory mediators in a subgroup of patients with chronic
obstructive pulmonary disease. Thorax 1996; 51:819-24.
111. Sauleda J, Garcia-Palmer FJ, Gonzalez G, Palou A, Agusti
AG. The activity of cytochrome oxidase is increased in cir-
culating lymphocytes of patients with chronic obstructive
pulmonary disease, asthma, and chronic arthritis. Am J
Respir Crit Care Med 2000; 161:32-5.
112. Sauleda J, Garcia-Palmer F, Wiesner RJ, Tarraga S,
Harting I, Tomas P, et al. Cytochrome oxidase activity and
mitochondrial gene expression in skeletal muscle of
patients with chronic obstructive pulmonary disease. Am J
Respir Crit Care Med 1998; 157:1413-7.
113. American Thoracic Society and European Respiratory
Society. Skeletal muscle dysfunction in chronic obstructive
pulmonary disease. Am J Respir Crit Care Med 1999;
159:S1-40.

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 43


GOLD_WR_05 8/18/05 12:56 PM Page 44

44 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


GOLD_WR_05 8/18/05 12:56 PM Page 45

CHAPTER

MANAGEMENT

OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 46

CHAPTER 5: MANAGEMENT OF COPD


INTRODUCTION
Management of Mild to Moderate COPD (Stages I and II) Patients should be identified as early in the course of the
involves the avoidance of risk factors to prevent disease disease as possible, and certainly before the end stage
progression and pharmacotherapy as needed to control of the illness when disability is substantial. However, the
symptoms. Severe (Stage III) and very severe (Stage IV) benefits of community-based spirometric screening, of
disease often require the integration of several different either the general population or smokers, are still unclear.
disciplines, a variety of treatment approaches, and a Educating patients and physicians to recognize that
commitment of the clinician to the continued support of cough, sputum production, and especially breathlessness
the patient as the illness progresses. In addition to are not trivial symptoms is an essential aspect of the
patient education, health advice, and pharmacotherapy, public health care of this disease.
COPD patients may require specific counseling about
smoking cessation, instruction in physical exercise, Reduction of therapy once symptom control has been
nutritional advice, and continued nursing support. Not all achieved is not normally possible in COPD. Further
approaches are needed for every patient, and assessing deterioration of lung function usually requires the
the potential benefit of each approach at each stage of progressive introduction of more treatments, both
the illness is a crucial aspect of effective disease pharmacologic and non-pharmacologic, to attempt to limit
management. the impact of these changes. Exacerbations of signs and
symptoms, a hallmark of COPD, impair patients' quality
An effective COPD management plan includes four of life and decrease their health status1,2. Appropriate
components: (1) Assess and Monitor Disease; (2) treatment and measures to prevent further exacerbations
Reduce Risk Factors; (3) Manage Stable COPD; (4) should be implemented as quickly as possible.
Manage Exacerbations.
Important differences exist between countries in the
While disease prevention is the ultimate goal, once approach to chronic illnesses such as COPD and in the
COPD has been diagnosed, effective management acceptability of particular forms of therapy. Ethnic
should be aimed at the following goals: differences in drug metabolism, especially for oral
medications, may result in different patient preferences
• Prevent disease progression. in different communities. Little is known about these
important issues in relationship to COPD.
• Relieve symptoms.
• Improve exercise tolerance.
REFERENCES
• Improve health status.
• Prevent and treat complications. 1. O'Brien C, Guest PJ, Hill SL, Stockley RA. Physiological and
• Prevent and treat exacerbations. radiological characterization of patients diagnosed with
chronic obstructive pulmonary disease in primary care.
• Reduce mortality.
Thorax 2000; 55:635-42.
2. Seemungal TA, Donaldson GC, Bhowmik A, Jeffries DJ,
These goals should be reached with minimal side effects Wedzicha JA. Time course and recovery of exacerbations in
from treatment, a particular challenge in COPD patients patients with chronic obstructive pulmonary disease. Am J
because they commonly have comorbidities. The extent Respir Crit Care Med 2000; 161:1608-13.
to which these goals can be realized varies with each
individual, and some treatments will produce benefits in
more than one area. In selecting a treatment plan, the
benefits and risks to the individual, and the costs, direct
and indirect, to the individual, his or her family, and the
community must be considered.

46 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 47

COMPONENT 1: ASSESS AND MONITOR DISEASE


can be caused by other lung diseases and by poor
performance. In the interest of improving the diagnosis of
KEY POINTS: COPD, every effort should be made to provide access to
standardized spirometry.
• Diagnosis of COPD is based on a history of
exposure to risk factors and the presence of Assessment of Symptoms
airflow limitation that is not fully reversible, with
or without the presence of symptoms. Although exceptions occur, the general patterns of
symptom development in COPD are well established.
• Patients who have chronic cough and sputum The main symptoms among patients in Stage 0: At Risk
production with a history of exposure to risk and Stage I: Mild COPD are chronic cough and sputum
factors should be tested for airflow limitation, production. These symptoms can be present for many
even if they do not have dyspnea. years before the development of airflow limitation and
are often ignored or discounted by patients. As airflow
• For the diagnosis and assessment of COPD, limitation develops in Stage II: Moderate COPD, patients
spirometry is the gold standard as it is the most often experience dyspnea, which may interfere with their
reproducible, standardized, and objective way of
measuring airflow limitation. FEV1/FVC < 70% Figure 5-1-1. Key Indicators for
and a postbronchodilator FEV1 < 80% predicted Considering a Diagnosis of COPD
confirms the presence of airflow limitation that is
not fully reversible. Consider COPD, and perform spirometry, if any of
these indicators are present. These indicators are not
• Health care workers involved in the diagnosis diagnostic by themselves, but the presence of multiple
and management of COPD patients should have
key indicators increases the probability of a diagnosis
access to spirometry.
of COPD. Spirometry is needed to establish a
diagnosis of COPD.
• Measurement of arterial blood gas tensions
should be considered in all patients with FEV1 < Chronic cough: Present intermittently or every day.
40% predicted or clinical signs suggestive of Often present throughout the day;
respiratory failure or right heart failure. seldom only nocturnal.

Chronic sputum Any pattern of chronic sputum


production: production may indicate COPD.
INITIAL DIAGNOSIS
Dyspnea that is: Progressive (worsens over time).
A diagnosis of COPD should be considered in any patient Persistent (present every day).
who has cough, sputum production, or dyspnea, and/or a Described by the patient as an
history of exposure to risk factors for the disease (Figure “increased effort to breathe,”
5-1-1). The diagnosis is confirmed by spirometry. The “heavi-ness,” “air hunger,” or “gasping.”
presence of a postbronchodilator FEV1 < 80% of the Worse on exercise.
predicted value in combination with an FEV1/FVC < 70% Worse during respiratory infections.
confirms the presence of airflow limitation that is not fully
reversible. Where spirometry is unavailable, the diagnosis History of Tobacco smoke.
of COPD should be made using all available tools. exposure to Occupational dusts and chemicals.
Clinical symptoms and signs, such as abnormal shortness risk factors, Smoke from home cooking and
of breath and increased forced expiratory time, can be especially: heating fuels.
used to help with the diagnosis. A low peak flow is
consistent with COPD, but has poor specificity since it

MANAGEMENT OF COPD 47
GOLD_WR_05 8/18/05 12:56 PM Page 48

daily activities. Typically, this is the stage at which they Figure 5-1-3. Questionnaire for Assessing the
seek medical attention and are diagnosed with COPD. Impact of Respiratory Symptoms6
However, some patients do not experience cough, spu-
tum production, or dyspnea in Stage I: Mild COPD or
Stage II: Moderate COPD, and do not come to medical WHEEZING
attention until their airflow limitation becomes more
severe or their lung function is worsened acutely by a Does your chest ever sound wheezing or whistling? Yes ❑
respiratory tract infection. As airflow limitation worsens No ❑
and the patient enters Stage III: Severe COPD, the symp-
toms of cough and sputum production typically continue, IF YOU ANSWERED “YES” TO THIS QUESTION:
dyspnea worsens, and additional symptoms heralding Do you get this on most days – or nights? Yes ❑
complications may develop. It is important to note that, No ❑
since COPD may be diagnosed at any stage, any of the
symptoms described below may be present in a patient Have you ever had attacks of shortness of breath Yes ❑
presenting for the first time. with wheezing? No ❑

IF YOU ANSWERED “YES” TO THIS QUESTION:


Is/was your breathing absolutely normal between Yes ❑
Figure 5-1-2. Causes of Chronic Cough
attacks? No ❑
with a Normal Chest X-ray
CHEST ILLNESSES
Intrathoracic
• Chronic obstructive pulmonary disease
During the last three years have you had any chest Yes ❑
• Bronchial asthma
illnesses which have kept you from your usual No ❑
• Central bronchial carcinoma
• Endobronchial tuberculosis activities for as much as a week?
• Bronchiectasis
• Left heart failure IF YOU ANSWERED YES TO THIS QUESTION:
• Interstitial lung disease Did you bring up phlegm more than usual during Yes ❑
• Cystic fibrosis these illnesses? No ❑

Extrathoracic IF YOU ANSWERED YES TO THIS QUESTION:


• Postnasal drip Have you had more than one illness like this in the Yes ❑
• Gastroesophageal reflux past three years? No ❑
• Drug therapy (e.g., ACE inhibitors)
BREATHLESSNESS

Cough. Chronic cough, usually the first symptom of PLEASE TICK IN THE BOX THAT APPLIES TO YOU
COPD to develop1, is often discounted by the patient as (ONE BOX ONLY)
an expected consequence of smoking and/or environ-
mental exposures. Initially, the cough may be intermit- I only get breathless with strenuous exercise. ❑
tent, but later is present every day, often throughout the
day, and is seldom entirely nocturnal. The chronic cough I get short of breath when hurrying on the level or ❑
in COPD may be unproductive2. In some cases, signifi- walking up a slight hill.
cant airflow limitation may develop without the presence
of a cough. Figure 5-1-2 lists some of the other causes I walk slower than people of the same age on the level ❑
of chronic cough in individuals with a normal chest X-ray. because of breathlessness, or I have to stop for breath
when walking on my own pace on the level.
Sputum production. COPD patients commonly raise
small quantities of tenacious sputum after coughing I stop for breath after walking about 100 yards or after a ❑
bouts. Regular production of sputum for 3 or more few minutes on the level.
months in 2 consecutive years is the epidemiological defi-
nition of chronic bronchitis3, but this is a somewhat arbi- I am too breathless to leave the house or I am breathless ❑
trary definition that does not reflect the range of sputum when dressing or undressing.
production in COPD patients. Sputum production is often

48 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 49

difficult to evaluate because patients may swallow sputum Hemoptysis can occur during respiratory tract infections
rather than expectorate it, a habit subject to significant in COPD patients9. However, this can be a sign of other
cultural and gender variation. diseases (e.g., tuberculosis, bronchial tumors) and
therefore should always be investigated. Cough syncope
Dyspnea. Dyspnea, the hallmark symptom of COPD, is occurs due to rapid increases in intrathoracic pressure
the reason most patients seek medical attention and is a during attacks of coughing. Coughing spells may also
major cause of disability and anxiety associated with the cause rib fractures, which are sometimes asymptomatic.
disease. Typical COPD patients describe their dyspnea Psychiatric morbidity, especially symptoms of depression
as a sense of increased effort to breathe, heaviness, air and/or anxiety, is common in advanced COPD10. Ankle
hunger, or gasping4. The terms used to describe dyspnea swelling can be the only symptomatic pointer to the
vary both by individual and by culture5. It is often possible development of cor pulmonale.
to distinguish the breathlessness of COPD from that due
to other causes by analysis of the terms used, although Medical History
there is considerable overlap with descriptors of bronchial
asthma. A simple way to quantify the impact of breath- A detailed medical history of a new patient known or
lessness on a patient’s health status is the British thought to have COPD should assess:
Medical Research Council (MRC) questionnaire (Figure
5-1-3). This questionnaire relates well to other measures • Patient’s exposure to risk factors, such as smoking
of health status6. and occupational or environmental exposures.

Breathlessness in COPD is characteristically persistent • Past medical history, including asthma, allergy,
and progressive. Even on “good days” COPD patients sinusitis or nasal polyps, respiratory infections in
experience dyspnea at lower levels of exercise than childhood, other respiratory diseases.
unaffected people of the same age. Initially, breathlessness • Family history of COPD or other chronic respiratory
is only noted on unusual effort (e.g., walking or running disease.
up a flight of stairs) and may be avoided entirely by
appropriate behavioral change (e.g., using an elevator). • Pattern of symptom development: COPD typically
As lung function deteriorates, breathlessness becomes develops in adult life and most patients are
more intrusive, and patients may notice that they are conscious of increased breathlessness, more
unable to walk at the same speed as other people of the frequent “winter colds,” and some social restriction
same age or carry out activities that require use of the for a number of years before seeking medical help.
accessory respiratory muscles (e.g., carrying grocery
bags)7. Eventually, breathlessness is present during • History of exacerbations or previous hospitalizations
everyday activities (e.g., dressing, washing) or at rest, for respiratory disorder: Patients may be aware of
leaving the patient confined to the home. periodic worsening of symptoms even if these
episodes have not been identified as exacerbations
of COPD.
Wheezing and chest tightness. Wheezing and chest
tightness are relatively non-specific symptoms that may • Presence of comorbidities, such as heart disease
vary between days, and over the course of a single day. and rheumatic disease, which may also contribute to
These symptoms may be present in Stage I: Mild COPD, restriction of activity.
but are more characteristic of asthma or Stage III:
Severe COPD and Stage IV: Very Severe COPD. Audible • Appropriateness of current medical treatments:
wheeze may arise at a laryngeal level and need not be For example, beta-blockers commonly prescribed
accompanied by ausculatory abnormalities. Alternatively, for heart disease are usually contraindicated in
widespread inspiratory or expiratory wheezes can be COPD.
present on listening to the chest. Chest tightness often
follows exertion, is poorly localized, is muscular in • Impact of disease on patient’s life, including limitation
character, and may arise from isometric contraction of the of activity; missed work and economic impact; effect
on family routines; feelings of depression or anxiety.
intercostal muscles. An absence of wheezing or chest
tightness does not exclude a diagnosis of COPD. • Social and family support available to the patient.

Additional symptoms in severe disease. Weight loss • Possibilities for reducing risk factors, especially
and anorexia are common problems in advanced COPD8. smoking cessation.

MANAGEMENT OF COPD 49
GOLD_WR_05 8/18/05 12:56 PM Page 50

Physical Examination
Figure 5-1-4. Considerations in
Though an important part of patient care, a physical Performing Spirometry
examination is rarely diagnostic in COPD. Physical signs
of airflow limitation are usually not present until significant
impairment of lung function has occurred11,12, and their Preparation
detection has a relatively low sensitivity and specificity. • Spirometers need calibration on a regular basis.
A number of physical signs may be present in COPD, but • Spirometers should produce hard copy to permit
their absence does not exclude the diagnosis. detection of technical errors.
• The supervisor of the test needs training in its effective
Inspection. performance.
• Maximal patient effort in performing the test is
• Central cyanosis, or bluish discoloration of the mucosal required to avoid errors in diagnosis and management.
membranes, may be present but is difficult to detect
in artificial light and in many racial groups. Performance
• Common chest wall abnormalities, which reflect the • Spirometry should be performed using techniques that
pulmonary hyperinflation seen in COPD, include meet published standards14.
relatively horizontal ribs, “barrel- shaped” chest, and • The expiratory volume/time traces should be smooth
protruding abdomen. and free from irregularities.
• Flattening of the hemi-diaphragms may be associated • The recording should go on long enough for a volume
with paradoxical in-drawing of the lower rib cage on plateau to be reached, which may take more than 12
inspiration, reduced cardiac dullness, and widening seconds in severe disease.
xiphisternal angle. • Both FVC and FEV1 should be the largest value
obtained from any of 3 technically satisfactory curves
• Resting respiratory rate is often increased to more
and the FVC and FEV1 values in these three curves
than 20 breaths per minute and breathing can be
should vary by no more than 5% or 100 ml, whichever
relatively shallow12.
is greater.
• Patients commonly show pursed-lip breathing, which
may serve to slow expiratory flow and permit more Evaluation
efficient lung emptying. • Spirometry measurements are evaluated by comparison
• COPD patients often have resting muscle activation of the results with appropriate reference values based
while lying supine. Use of the scalene and stern- on age, height, sex, and race (e.g., see reference 14).
ocleidomastoid muscles is a further indicator of • The presence of a postbronchodilator FEV1 < 80%
respiratory distress. predicted together with an FEV1/FVC < 70% confirms
the presence of airflow limitation that is not fully
• Ankle or lower leg edema can be a sign of right heart
reversible.
failure.
• In patients with FEV1 > 80% predicted, FEV1/FVC <
70% may be an early indicator of developing airflow
Palpation and percussion. limitation.

• These are often unhelpful in COPD. • The presence of wheezing during quiet breathing is a
useful pointer to airflow limitation. However, wheezing
• Detection of the heart apex beat may be difficult due
heard only after forced expiration is of no diagnostic
to pulmonary hyperinflation.
value.
• Hyperinflation also leads to downward displacement
• Inspiratory crackles occur in some COPD patients but
of the liver and an increase in the ability to palpate are of little help diagnostically.
this organ without it being enlarged.
• Heart sounds are best heard over the xiphoid area.

Auscultation.
Measurement of Airflow Limitation
(Spirometry)
• Patients with COPD often have reduced breath
sounds, but this finding is not sufficiently characteristic Spirometry measurements should be undertaken for any
to make the diagnosis13. patient who may have COPD. To help identify individuals
earlier in the course of the disease, spirometry should be

50 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 51

performed for patients who have chronic cough and the presence of airflow limitation that is not fully
sputum production even if they do not have dyspnea. reversible. The FEV1/FVC on its own is a more sensitive
Although spirometry does not fully capture the impact of measure of airflow limitation, and an FEV1/FVC < 70% is
COPD on a patient’s health, it remains the gold standard
considered an early sign of airflow limitation in patients
for diagnosing the disease and monitoring its progression.
whose FEV1 remains normal (≥ 80% predicted). This
It is the best standardized, most reproducible, and most
approach to defining airflow limitation is a pragmatic one
objective measurement of airflow limitation available.
in view of the fact that universally applicable reference
Health care workers who care for COPD patients should
have access to spirometry, which is useful in both diagnosis values for FEV1 and FVC are not available.
and periodic monitoring. Figure 5-1-4 summarizes some
considerations that are crucial to achieving consistently Peak expiratory flow (PEF) is sometimes used as a measure
accurate test results. of airflow limitation, but in COPD the relationship between
PEF and FEV1 is poor. PEF may underestimate the
degree of airways obstruction in these patients15. If
Spirometry should measure the maximal volume of air spirometry is unavailable, prolongation of the forced
forcibly exhaled from the point of maximal inspiration expiratory time beyond 6 seconds is a crude, but useful,
(forced vital capacity, FVC) and the volume of air exhaled guide to the presence of an FEV1/FVC ratio < 50%16,17.
during the first second of this maneuver (forced expiratory
volume in one second, FEV1), and the ratio of these two The role of screening spirometry in the general population
measurements (FEV1/FVC) should be calculated. or in a population at risk for COPD is controversial. Both
Spirometry measurements are evaluated by comparison FEV1 and FVC predict all-cause mortality independent of
with reference values based on age, height, sex, and race tobacco smoking, and abnormal lung function identifies a
(use appropriate reference values, e.g., see reference 14). subgroup of smokers at increased risk for lung cancer.
This has been the basis of an argument that screening
Figure 5-1-5 shows a normal spirogram and a spirogram spirometry should18
be employed as a global health
typical of patients with mild to moderate COPD. Patients assessment tool . However, there are no data to indicate
with COPD typically show a decrease in both FEV1 and that screening spirometry is effective in directing
FVC. The degree of spirometric abnormality generally management decisions or in improving COPD outcomes.
reflects the severity of COPD (Figure 1-2). The presence
of a postbronchodilator FEV1 < 80% of the predicted Assessment of Severity
value in combination with an FEV1/FVC < 70% confirms
Assessment of COPD severity is based on the patient’s
level of symptoms, the severity of the spirometric
Figure 5-1-5. Normal Spirogram and Spirogram abnormality, and the presence of complications such as
Typical of Patients with Moderate COPD respiratory failure and right heart failure (Figure 1-2).
The use of specific spirometric cut-points (e.g.,
FEV1/FVC <70% and FEV1 < 80% predicted) to define
different stages of COPD is for purposes of simplicity;
these cut-points have not been clinically validated and
FEV1 FVC FEV1/FVC may overestimate the prevalence of COPD in some
groups, such as the elderly.

Although the presence of airflow limitation is key to the


current understanding of COPD, it may be valuable from
FEV1 a public health perspective to identify individuals at risk
Liters

for the disease before significant airflow limitation develops


(Stage 0, At Risk). A majority of people with early COPD
identified in large studies complained of at least one
FEV1 respiratory symptom, such as cough, sputum production,
wheezing, or breathlessness19,20. These symptoms may
be present at a time of relatively minor or even no spiro-
metric abnormality. While not all individuals
21
with such
symptoms will go on to develop COPD , the presence of
these symptoms should help define a high-risk population

MANAGEMENT OF COPD 51
GOLD_WR_05 8/18/05 12:56 PM Page 52

that should be targeted for preventive intervention. Much


depends on the success of convincing such people, as Figure 5-1-6. Bronchodilator Reversibility Testing
well as health care workers, that minor respiratory
symptoms may be markers of future ill health. Preparation

• Tests should be performed when patients are clinically


The severity of a patient’s breathlessness is important stable and free from respiratory infection.
and can be gauged by the MRC scale (Figure 5-1-3).
Arterial blood gases should be measured in all patients • Patients should not have taken inhaled short-acting
who have FEV1 < 40% predicted or clinical signs of bronchodilators in the previous six hours, long-acting
respiratory failure or right heart failure. ß2 agonists in the previous 12 hours, or sustained-
release theophyllines in the previous 24 hours.
Additional Investigations
Spirometry
For patients diagnosed with Stage II: Moderate COPD
and beyond, the following additional investigations may • FEV1 should be measured before a bronchodilator is
be useful. given.

• The bronchodilator should be given by metered dose


Bronchodilator reversibility testing. Generally performed
only once, at the time of diagnosis, this test is useful for inhaler through a spacer device or by nebulizer to be
several reasons: certain it has been inhaled.

• The bronchodilator dose should be selected to be


• To help rule out a diagnosis of asthma. high on the dose/response curve.
If FEV1 returns to the predicted normal range after
administration of a bronchodilator, the patient’s airflow • Suitable dosage protocols are 400 g ß2-agonist,
limitation is likely due to asthma. 80 g anticholinergic, or the two combined. FEV1
should be measured again 30-45 minutes after the
• To establish a patient’s best attainable lung function at bronchodilator is given.
that point in time.

• To gauge a patient’s prognosis. Results


Some studies show that the postbronchodilator FEV1 is • An increase in FEV1 that is both greater than 200 ml
a more reliable prognostic marker than pre-bronchodilator
and 12% above the pre-bronchodilator FEV1 is con-
FEV122. In addition, the Intermittent Positive Pressure
Breathing (IPPB) Study, a multicenter clinical trial, sidered significant.
suggested that the degree of bronchodilator response
is inversely related to the rate of FEV1 decline in
COPD patients23. Between-day reproducibility of spirometry in the same
individual is approximately 178 ml24. Thus, an acute
• To assess potential response to treatment. change that exceeds both 200 ml and 12% of the base-
Patients who show significant improvement in FEV1 line measurement is unlikely to have arisen by chance.
after administration of a bronchodilator are more likely A protocol for bronchodilator reversibility testing is listed
to benefit from treatment with bronchodilators and have in Figure 5-1-6.
a positive response to glucocorticosteroids. However,
individual responses to bronchodilator tests are influenced
by many factors, and failure of FEV1 to change by an
arbitrary amount on one day does not preclude a Chest X-ray. A chest X-ray is seldom diagnostic in
response on another. Moreover, even patients who do COPD unless obvious bullous disease is present, but it is
not show a significant FEV1 response to a short-acting valuable in excluding alternative diagnoses. Radiological
bronchodilator test may benefit symptomatically from changes associated with COPD include signs of hyperin-
long-term bronchodilator therapy. flation (flattened diaphragm on the lateral chest film, and
an increase in the volume of the retrosternal air space),

52 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 53

hyperlucency of the lungs, and rapid tapering of the vascular


Figure 5-1-7. Differential Diagnosis of COPD markings. Computed tomography (CT) of the chest is not
routinely recommended. However, when there is doubt
Diagnosis Suggestive Features* about the diagnosis of COPD, high resolution CT (HRCT)
might help in the differential diagnosis. In addition, if a
COPD Onset in mid-life. surgical procedure such as bullectomy or lung volume
Symptoms slowly progressive. reduction is contemplated, chest CT is helpful.
Long smoking history.
Dyspnea during exercise. Arterial blood gas measurement. In advanced COPD
Largely irreversible airflow limitation. measurement of arterial blood gases is important. This
Asthma Onset early in life (often childhood). test should be performed in patients with FEV1 < 40%
Symptoms vary from day to day. predicted or with clinical signs suggestive of respiratory
Symptoms at night/early morning. failure or right heart failure.
Allergy, rhinitis, and/or eczema also
present. Alpha-1 antitrypsin deficiency screening. In patients
Family history of asthma. who develop COPD at a young age (< 45 years) or who
Largely reversible airflow limitation. have a strong family history of the disease, it may be
valuable to identify coexisting alpha-1 antitrypsin deficiency.
Congestive Fine basilar crackles on auscultation.
This could lead to family screening or appropriate
Heart Failure Chest X-ray shows dilated heart,
counseling. A serum concentration of alpha-1 antitrypsin
pulmonary edema.
below 15-20 % of the normal value is highly suggestive
Pulmonary function tests indicate
of homozygous alpha-1 antitrypsin deficiency.
volume restriction, not airflow limitation.
Bronchiectasis Large volumes of purulent sputum. Differential Diagnosis
Commonly associated with bacterial
infection. A major differential diagnosis is asthma. In some patients
Coarse crackles/clubbing on with chronic asthma, a clear distinction from COPD is not
auscultation. possible using current imaging and physiological testing
Chest X-ray/CT shows bronchial techniques, and it is assumed that asthma and COPD
dilation, bronchial wall thickening. coexist in these patients. In these cases, current
Tuberculosis Onset all ages. management is similar to that of asthma. Other potential
Chest X-ray shows lung infiltrate. diagnoses are usually easier to distinguish from COPD
Microbiological confirmation. (Figure 5-1-7).
High local prevalence of tuberculosis.
Obliterative Onset in younger age, nonsmokers. ONGOING MONITORING
Bronchiolitis May have history of rheumatoid AND ASSESSMENT
arthritis or fume exposure.
CT on expiration shows hypodense
Visits to health care facilities will increase in frequency
areas.
as COPD progresses. The type of health care workers
Diffuse Most patients are male and non- seen, and the frequency of visits, will depend on the
Panbronchiolitis smokers. health care system. Ongoing monitoring and assessment
Almost all have chronic sinusitis. in COPD ensures that the goals of treatment are being
Chest X-ray and HRCT show diffuse met and should include evaluation of: (1) exposure to
small centrilobular nodular opacities risk factors, especially tobacco smoke; (2) disease
and hyperinflation. progression and development of complications;
(3) pharmacotherapy and other medical treatment;
(4) exacerbation history; (5) comorbidities.
*These features tend to be characteristic of the respective diseases,
but do not occur in every case. For example, a person who has
never smoked may develop COPD (especially in the developing Suggested questions for follow-up visits are listed in
world, where other risk factors may be more important than cigarette Figure 5-1-8. The best way to detect changes in
smoking); asthma may develop in adult and even elderly patients.
symptoms and overall health status is to ask the same
questions at each visit.

MANAGEMENT OF COPD 53
GOLD_WR_05 8/18/05 12:56 PM Page 54

Monitor Disease Progression and Figure 5-1-8. Suggested Questions


Development of Complications for Follow-Up Visits*

COPD is usually a progressive disease. Lung function Monitor exposure to risk factors:
can be expected to worsen over time, even with the best • Have you continued to stay off cigarettes?
available care. Symptoms and objective measures of • If not, how many cigarettes per day are you smoking?
airflow limitation should be monitored to determine when • Would you like to quit smoking?
to modify therapy and to identify any complications that • Has there been any change in your working
may develop. As at the initial assessment, follow-up environment?
visits should include a physical examination and
discussion of symptoms, particularly any new or Monitor disease progression and development
worsening symptoms. of complications:
• How much can you do before you get short of breath?
Pulmonary function. A patient’s decline in lung function (Use an everyday example, such as walking up flights
is best tracked by periodic spirometry measurements. of stairs, up a hill, or on flat ground.)
Useful information about lung function decline is unlikely • Has your dyspnea worsened, improved, or stayed the
from spirometry measurements performed more than same since your last visit?
once a year. Spirometry should be performed if there is • Have you had to reduce your activities because of
a substantial increase in symptoms or a complication. dyspnea or other symptoms?
• Have any of your symptoms worsened since your
Other pulmonary function tests, such as flow-volume last visit?
loops, diffusing capacity (DLCO) measurements, and • Have you experienced any new symptoms since your
measurement of lung volumes are not needed in a last visit?
routine assessment but can provide information about the • Has your sleep been disrupted due to dyspnea or other
overall impact of the disease and can be valuable in symptoms?
resolving diagnostic uncertainties and assessing patients • Since your last visit, have you missed any work
for surgery. because of your symptoms?

Arterial blood gas measurement. Measurement of Monitor pharmacotherapy and other medical
arterial blood gas tensions should be performed in all treatment:
patients with FEV1 < 40% predicted or when clinical • What medications are you taking?
signs of respiratory failure or right heart failure are • How often do you take each medication?
present. Respiratory failure is indicated by a • How much do you take each time?
PaO2 < 8.0 kPa (60 mm Hg) with or without • Have you missed or stopped taking any regular doses
PaCO2 > 6.7 kPa (50 mm Hg) in arterial blood gas of your medications for any reason?
measurements made while breathing air at sea level. • Have you had trouble filling your prescriptions
(e.g., for financial reasons, not on formulary)?
Screening patients by pulse oximetry and assessing • Please show me how you use your inhaler.
arterial blood gases in those with an oxygen saturation • Have you tried any other medicines or remedies?
(SaO2) < 92% may be a useful way of selecting patients • Has your medication been effective in controlling
for arterial blood gas measurement27. However, pulse your symptoms?
oximetry gives no information about CO2 tensions. • Has your medication caused you any problems?

Several considerations are important to ensure accurate Monitor exacerbation history:


test results. Oxygen pressure in the inspired air (FiO2) • Since your last visit, have you had any episodes/times
should be measured, taking note if patient is using an when your symptoms were a lot worse than usual?
O2-driven nebulizer. Changes in arterial blood gas ten- • If so, how long did the episode(s) last? What do you
sions take time to occur, especially in severe disease. think caused the symptoms to get worse? What did you
Thus, 20-30 minutes should pass before rechecking the do to control the symptoms?
gas tensions when the FiO2 has been changed.
*These questions are examples and do not represent a standardized
assessment instrument. The validity and reliability of these questions
have not been assessed.

54 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 55

Adequate pressure must be applied at the puncture site Sleep studies. Sleep studies may be indicated when
for at least one minute; failure to do so can lead to painful hypoxemia or right heart failure develops in the presence
bruising. of relatively mild airflow limitation or when the patient has
symptoms suggesting the presence of sleep apnea.
Clinical signs of respiratory failure or right heart failure
include central cyanosis, ankle swelling, and an increase Exercise testing. Several types of tests are available to
in the jugular venous pressure. Clinical signs of hyper- measure exercise capacity, but these are primarily used
capnia are extremely nonspecific outside of exacerbations. in conjunction with pulmonary rehabilitation programs.

Assessment of pulmonary hemodynamics. Pulmonary Monitor Pharmacotherapy and Other


hypertension is only likely to be important in patients who Medical Treatment
have developed respiratory failure. Measurement of pul-
monary arterial pressure is not recommended in clinical In order to adjust therapy appropriately as the disease
practice as it does not add practical information beyond progresses, each follow-up visit should include a discussion
that obtained from a knowledge of PaO2. of the current therapeutic regimen. Dosages of various
medications, adherence to the regimen, inhaler technique,
Diagnosis of right heart failure or cor pulmonale. effectiveness of the current regime at controlling symptoms,
Elevation of the jugular venous pressure and the pres- and side effects of treatment should be monitored.
ence of pitting ankle edema are often the most useful
findings suggestive of cor pulmonale in clinical practice. Monitor Exacerbation History
However, the jugular venous pressure is often difficult to
assess in patients with COPD, due to large swings in During periodic assessments, health care workers should
intrathoracic pressure. Firm diagnosis of cor pulmonale question the patient and evaluate any records of exacer-
can be made through a number of investigations, includ- bations, both self-treated and those treated by other
ing radiography, electrocardiography, echocardiography, health care providers. Frequency, severity, and likely
radionucleotide scintigraphy, and magnetic resonance causes of exacerbations should be evaluated. Increased
imaging. However, all of these measures involve inherent sputum volume, acutely worsening dyspnea, and the
inaccuracies of diagnosis. presence of purulent sputum should be noted. Specific
inquiry into unscheduled visits to providers, telephone
CT and ventilation-perfusion scanning. Despite the calls for assistance, and use of urgent or emergency care
benefits of being able to delineate pathological anatomy, facilities may be helpful. Severity can be estimated by
routine CT and ventilation-perfusion scanning are currently the increased need for bronchodilator medication or glu-
confined to the assessment of COPD patients for surgery. cocorticosteroids and by the need for antibiotic treatment.
HRCT is currently under investigation as a way of visual- Hospitalizations should be documented, including the
izing airway and parenchymal pathology more precisely. facility, duration of stay, and any use of critical care or
intubation. The clinician then can request summaries of
Hematocrit. Polycythemia can develop in the presence all care received to facilitate continuity of care.
of arterial hypoxemia, especially in continuing smokers28.
Polycythemia can be identified by hematocrit > 55%. Monitor Comorbidities

Respiratory muscle function. Respiratory muscle func- In treating patients with COPD, it is important to consider
tion is usually measured by recording the maximum inspi- the presence of concomitant conditions such as bronchial
ratory and expiratory mouth pressures. More complex carcinoma, tuberculosis, sleep apnea, and left heart fail-
measurements are confined to research laboratories. ure. The appropriate diagnostic tools (chest radiograph,
Measurement of expiratory muscle force is useful in ECG, etc.) should be used whenever symptoms (e.g.,
assessing patients when dyspnea or hypercapnia is not hemoptysis) suggest one of these conditions.
readily explained by lung function testing or when periph-
eral muscle weakness is suspected. This measurement
may improve in COPD patients when other measure-
ments of lung mechanics do not (e.g., after pulmonary
rehabilitation)29,30.

MANAGEMENT OF COPD 55
GOLD_WR_05 8/18/05 12:56 PM Page 56

REFERENCES 15. Kelly CA, Gibson GJ. Relation between FEV1 and peak
expiratory flow in patients with chronic obstructive pul-
1. Georgopoulos D, Anthonisen NR. Symptoms and signs of monary disease. Thorax 1988; 43:335-6.
COPD. In: Cherniack NS, ed. Chronic obstructive pul- 16. Lal S, Ferguson AD, Campbell EJM. Forced expiratory time;
monary disease. Toronto: WB Saunders; 1991. p. 357-63. a simple test for airways obstruction. BMJ 1964; 1:814-7.
2. Burrows B, Niden AH, Barclay WR, Kasik JE. Chronic 17. Swanney MP, Jensen RL, Crichton DA, Beckert LE, Cardno
obstructive lung disease II. Relationships of clinical and LA, Crapo RO. FEV(6) is an acceptable surrogate for FVC
physiological findings to the severity of airways obstruction. in the spirometric diagnosis of airway obstruction and
Am Rev Respir Dis 1965; 91:665-78. restriction. Am J Respir Crit Care Med 2000; 162:917-9.
3. Medical Research Council. Definition and classification of 18. Ferguson GT, Enright PL, Buist AS, Higgins MW. Office
chronic bronchitis for clinical and epidemiological purpos- spirometry for lung health assessment in adults: a consen-
es: a report to the Medical Research Council by their sus statement from the national lung health education pro-
Committee on the Aetiology of Chronic Bronchitis. Lancet gram. Chest 2000; 117:1146-61.
1965; 1:775-80. 19. Kanner RE, Connett JE, Williams DE, Buist AS. Effects of
4. Simon PM, Schwartstein RM, Weiss JW, Fencl V, randomized assignment to a smoking cessation interven-
Teghtsoonian M, Weinberger SE. Distinguishable types of tion and changes in smoking habits on respiratory symp-
dyspnea in patients with shortness of breath. Am Rev toms in smokers with early chronic obstructive pulmonary
Respir Dis 1990; 142:1009-14. disease: the Lung Health Study. Am J Med 1999; 106:410-6.
5. Elliott MW, Adams L, Cockcroft A, MacRae KD, Murphy K, 20. Lofdahl CG, Postma DS, Laitinen LA, Ohlsson SV, Pauwels
Guz A. The language of breathlessness. Use of verbal RA, Pride NB. The European Respiratory Society study on
descriptors by patients with cardiopulmonary disease. Am chronic obstructive pulmonary disease (EUROSCOP):
Rev Respir Dis 1991; 144:826-32. recruitment methods and strategies. Respir Med 1998;
6. Bestall JC, Paul EA, Garrod R, Garnham R, Jones PW, 92:467-72.
Wedzicha JA. Usefulness of the Medical Research 21. Peto R, Speizer FE, Cochrane AL, Moore F, Fletcher CM,
Council (MRC) dyspnoea scale as a measure of disability Tinker CM, et al. The relevance in adults of airflow obstruc-
in patients with chronic obstructive pulmonary disease. tion, but not of mucus hypersecretion, to mortality from
Thorax 1999; 54:581-6. chronic lung disease: results from twenty years of prospec-
7. Celli BR, Rassulo J, Make BJ. Dyssynchronous breathing tive observation. Am Rev Respir Dis 1983; 128:491-500.
during arm but not leg exercise in patients with chronic air- 22. Hansen EF, Phanareth K, Laursen LC, KokJensen A,
flow obstruction. N Engl J Med 1986; 314:1485-90. Dirksen A. Reversible and irreversible airflow obstruction
8. Schols AM, Soeters PB, Dingemans AM, Mostert R, as predictor of overall mortality in asthma and chronic
Frantzen PJ, Wouters EF. Prevalence and characteristics of obstructive pulmonary disease. Am J Respir Crit Care
nutritional depletion in patients with stable COPD eligible Med 1999; 159:1267-71.
for pulmonary rehabilitation. Am Rev Respir Dis 1993; 23. Anthonisen NR, Wright EC. Bronchodilator response in
147:1151-6. chronic obstructive pulmonary disease. Am Rev Respir
9. Johnston RN, Lockhart W, Ritchie RT, Smith DH. Dis 1986; 133:814-9.
Haemoptysis. BMJ 1960; 1:592-5. 24. Sourk RL, Nugent KM. Bronchodilator testing: confidence
10. Calverley PM. Neuropsychological deficits in chronic intervals derived from placebo inhalations. Am Rev Respir
obstructive pulmonary disease. Monaldi Archives for Chest Dis 1983; 128:153-7.
Disease 1996; 51:5-6. 25. Reis AL. Response to bronchodilators. In: Clausen J, ed.
11. Kesten S, Chapman KR. Physician perceptions and man- Pulmonary function testing: guidelines and controversies.
agement of COPD. Chest 1993; 104:254-8. New York: Academic Press; 1982. p. 215-221.
12. Loveridge B, West P, Kryger MH, Anthonisen NR. Alteration 26. American Thoracic Society. Lung function testing: selection
of breathing pattern with progression of chronic obstruc- of reference values and interpretative strategies. Am Rev
tive pulmonary disease. Am Rev Respir Dis 1986; 134:930-4. Respir Dis 1991; 144:1202-18.
13. Badgett RC, Tanaka DV, Hunt DK, Jelley MJ, Feinberg LE, 27. Roberts CM, Bugler JR, Melchor R, Hetzel ML, Spiro SG.
Steiner JF, et al. Can moderate chronic obstructive pul- Value of pulse oximetry for long-term oxygen therapy
monary disease be diagnosed by history and physical find- requirement. Eur Respir J 1993; 6:559-62.
ings alone? Am J Med 1993; 94:188-96. 28. Calverley PM, Leggett RJ, McElderry L, Flenley DC.
14. Standardization of spirometry, 1994 update. Am J Respir Cigarette smoking and secondary polycythemia in hypoxic
Crit Care Med 1995; 152:1107-36. cor pulmonale. Am Rev Respir Dis 1982; 125:507-10.

56 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 57

29. Dekhuijzen PNR, Folgering HT, van Herwaarden CLA.


Target-flow inspiratory muscle training during pulmonary
rehabilitation in patients with COPD. Chest 1991; 99:128-33.
30. Heijdra YF, Dekhuijzen PN, van Herwaarden CLA,
Forlgering H. Nocturnal saturation improves by target-flow
inspiratory muscle training in patients with COPD. Am J
Respir Crit Care Med 1996; 153:260-5.

MANAGEMENT OF COPD 57
GOLD_WR_05 8/18/05 12:56 PM Page 58

COMPONENT 2: REDUCE RISK FACTORS


implemented and smoking cessation programs should
KEY POINTS: be readily available and encouraged for all individuals
who smoke. Reduction of total personal exposure to
• Reduction of total personal exposure to tobacco occupational dust, fumes, and gases and to indoor and
smoke, occupational dusts and chemicals, and outdoor air pollutants is also an important goal to prevent
indoor and outdoor air pollutants are important the onset and progression of COPD1.
goals to prevent the onset and progression of
COPD. TOBACCO SMOKE
• Smoking cessation is the single most effective - Smoking Prevention
and cost effective - way in most people to reduce
the risk of developing COPD and stop its progression Comprehensive tobacco control policies and programs
(Evidence A). with clear, consistent, and repeated nonsmoking
messages should be delivered through every feasible
• Brief tobacco dependence counseling is effective channel, including health care providers, schools, and
(Evidence A) and every tobacco user should be radio, television, and print media. National and local
offered at least this treatment at every visit to a campaigns should be undertaken to reduce exposure to
health care provider. tobacco smoke in public forums. Legislation to establish
smoke-free schools, public facilities, and work environments
• Three types of counseling are especially effective: should be encouraged by government officials, public
practical counseling, social support as part of health workers, and the public. Smoking prevention
treatment, and social support arranged outside of programs should target all ages, including young children,
treatment (Evidence A). adolescents, young adults, and pregnant women.
Physicians and public health officials should encourage
• Several effective pharmacotherapies for tobacco smoke-free homes.
dependence are available (Evidence A), and at
least one of these medications should be added The first exposure to cigarette smoke may begin in utero
to counseling if necessary and in the absence of when the fetus is exposed to blood-borne metabolites
contraindications (Evidence A). from the mother2,3. Neonates and infants may be
exposed passively to tobacco smoke in the home if a
• Progression of many occupationally induced family member smokes. Children less than 2 years old
respiratory disorders can be reduced or controlled who are passively exposed to cigarette smoke have an
through a variety of strategies aimed at reducing increased prevalence of respiratory infections, and are at
the burden of inhaled particles and gases a greater risk of developing chronic respiratory symptoms
(Evidence B). later in life4.

Smoking Cessation

Smoking cessation is the single most effective - and cost


INTRODUCTION effective - way to reduce exposure to COPD risk factors.
Quitting smoking can prevent or delay the development of
Identification, reduction, and control of risk factors are airflow limitation or reduce its progression5. A statement
important steps toward prevention and treatment of any by the WHO (Figure 5-2-1)6 emphasizes the health and
disease. In the case of COPD, these factors include economic benefits to be gained from smoking cessation.
tobacco smoke, occupational exposures, and indoor and All smokers - including those who may be at risk for
outdoor air pollution and irritants. COPD as well as those who already have the disease -
should be offered the most intensive smoking cessation
Since cigarette smoking is the major risk factor for COPD intervention feasible.
worldwide, smoking prevention programs should be

58 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 59

The role of health care providers in smoking cessation.


Figure 5-2-1. World Health Organization Statement A successful smoking cessation strategy requires a
on Smoking Cessation6 multifaceted approach, including public policy, information
dissemination programs, and health education through
Smoking cessation is a critical step toward substantially the media and schools9. However, health care providers,
reducing the health risks run by current smokers, including physicians, nurses, dentists, psychologists,
thereby improving world health. Tobacco has been pharmacists, and others, are key to the delivery of smoking
shown to cause about 25 life-threatening diseases, cessation messages and interventions. Involving as
or groups of diseases, many of which can be prevented, many of these individuals as possible will help. Health
delayed, or mitigated by smoking cessation. As life care workers should encourage all patients who smoke
expectancy increases in developing countries, the to quit, even those patients who come to the health care
morbidity and mortality burden of chronic diseases provider for unrelated reasons and do not have symptoms
will increase still further. This projected concentration of COPD or evidence of airflow limitation.
of tobacco-related disease burden can be lightened
by intensive efforts at smoking cessation. Studies Guidelines for smoking cessation entitled Treating
have shown that 75-80% of smokers want to quit, Tobacco Use and Dependence: A Clinical Practice
while one-third have made at least three serious Guideline were published by the US Public Health
cessation attempts. Cessation efforts cannot be Service10. The major conclusions are summarized in
ignored in favor of primary prevention; rather, both Figure 5-2-2.
efforts must be made in conjunction with one another.
If only small portions of today's 1.1 billion smokers
were able to stop, the long-term health and economic Figure 5-2-2. Public Health Service Report: Treating
benefits would be immense. Governments, Tobacco Use and Dependence: A Clinical Practice
communities, organizations, schools, families and Guideline - Major Findings and Recommendations10
individuals are called upon to help current smokers
stop their addictive and damaging habit. 1. Tobacco dependence is a chronic condition that
warrants repeated treatment until long-term or
permanent abstinence is achieved.
2. Effective treatments for tobacco dependence exist
Smoking cessation interventions are effective in both and all tobacco users should be offered these
genders, in all racial and ethnic groups, and in pregnant treatments.
women. Age influences quit rates, with young people 3. Clinicians and health care delivery systems
less likely to quit, but nevertheless smoking cessation must institutionalize the consistent identification,
programs can be effective in all age groups. documentation and treatment of every tobacco
user at every visit.
International data on the economic impact of smoking 4. Brief tobacco dependence treatment is effective
cessation are strikingly consistent: investing resources in and every tobacco user should be offered at least
brief treatment.
smoking cessation programs is cost effective in terms of
medical costs per life year gained. Interventions that 5. There is a strong dose-response relation between the
have been investigated include nicotine replacement with intensity of tobacco dependence counseling and its
effectiveness.
transdermal patch, counseling from physicians and other
health professionals (with and without nicotine patch), 6. Three types of counseling were found to be especially
self-help and group programs, and community-based effective: practical counseling, social support as part
of treatment, and social support arranged outside of
stop-smoking contests. A review of data from a number
treatment.
of countries estimated the median societal cost of various
smoking cessation interventions at $990 to $13,000 (US) 7. Five first-line pharmacotherapies for tobacco dependence
- bupropion SR, nicotine gum, nicotine inhaler, nicotine
per life year gained7. Smoking cessation programs are a
nasal spray, and nicotine patch - are effective and at
particularly good value for the UK National Health least one of these medications should be prescribed
Service, with costs from £212 to £873 (US $320 to in the absence of contraindications.
$1,400) per life year gained8.
8. Tobacco dependence treatments are cost effective
relative to other medical and disease prevention
interventions.

MANAGEMENT OF COPD 59
GOLD_WR_05 8/18/05 12:56 PM Page 60

The Public Health Service Guidelines recommend a


Figure 5-2-3. Brief Strategies to Help the
five-step program for intervention (Figure 5-2-3), which
Patient Willing to Quit10-13
provides a strategic framework helpful to health care
providers interested in helping their patients stop
1. ASK: Systematically identify all tobacco users at smoking10-13. The guidelines emphasize that tobacco
every visit. dependence is a chronic disease (Figure 5-2-4)10 and
Implement an office-wide system that ensures that, urge clinicians to recognize that relapse is common and
for EVERY patient at EVERY clinic visit, tobacco-use reflects the chronic nature of dependence, not failure on
status is queried and documented. the part of the clinician or the patient.

2. ADVISE: Strongly urge all tobacco users to quit.


Figure 5-2-5.
In a clear, strong, and personalized manner, urge
Stages of Change Model
every tobacco user to quit.

3. ASSESS: Determine willingness to make a quit Precontemplation


attempt.
Ask every tobacco user if he or she is willing to make
a quit attempt at this time (e.g., within the next 30 days). Relapse Contemplation Preparation

4. ASSIST: Aid the patient in quitting.


Slipping Back Recycling Action
Help the patient with a quit plan; provide practical
counseling; provide intra-treatment social support; help the
patient obtain extra-treatment social support; recommend Short-Term
use of approved pharmacotherapy except in special Maintenance
circumstances; provide supplementary materials.
Sustained
5. ARRANGE: Schedule follow-up contact. Maintenance
Schedule follow-up contact, either in person or via
telephone. Printed with permission of Dr. Peter M.A. Calverley.

Most individuals go through several stages before they


Figure 5-2-4. Tobacco Dependence stop smoking (Figure 5-2-5)9. It is often helpful for the
as a Chronic Disease10 clinician to assess a patient's readiness to quit in order to
determine the most effective course of action at that time.
• For most people, tobacco dependence results in The clinician should initiate treatment if the patient is
true drug dependence comparable to dependence ready to quit. For a patient not ready to make a quit
caused by opiates, amphetamines, and cocaine. attempt, the clinician should provide a brief intervention
designed to promote the motivation to quit.
• Tobacco dependence is almost always a chronic
disorder that warrants long-term clinical intervention Counseling. Counseling delivered by physicians and
as do other addictive disorders. Failure to appreciate other health professionals significantly increases quit
the chronic nature of tobacco dependence may rates over self-initiated strategies14. Even a brief
impair the clinician's motivation to treat tobacco use (3-minute) period of counseling to urge a smoker to quit
consistently in a long-term fashion. results in smoking cessation rates of 5-10%15. At the very
least, this should be done for every smoker at every
• Clinicians must understand that this is a chronic health care provider visit15,16.
condition comparable to diabetes, hypertension,
or hyperlipidemia requiring simple counseling advice, However, there is a strong dose-response relationship
support, and appropriate pharmacotherapy. between counseling intensity and cessation success17,18.
Ways to make the treatment more intense include
• Relapse is common, which is the nature of dependence increasing the length of the treatment session, the
and not the failure of the clinician or the patient. number of treatment sessions, and the number of weeks
over which the treatment is delivered. Counseling

60 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 61

sessions of 3 to 10 minutes result in cessation rates of Figure 5-2-6. Patient Support in


around 12%10. With more complex interventions (for Smoking Cessation Programs9,10
example, controlled clinical trials that include skills training,
problem solving, and psychosocial support), quit rates can • Encourage the patient in the quit attempt.
reach 20-30%17. In one multicenter controlled clinical trial, Indicate that effective cessation treatments are now
a combination of physician advice, group support, skills available and, in fact, half of all people who smoked
training, and nicotine replacement therapy achieved a have now quit. Communicate your confidence in the
quit rate of 35% at one year and a sustained quit rate of patient's ability to quit.
22% at 5 years5.
• Communicate care and concern. Ask how the
Both individual and group counseling are effective formats patient feels about smoking and whether he/she
for smoking cessation. Several particular items of wants to quit, expressing concern along with the
counseling content seem to be especially effective, ability and willingness to help. Be open to the
including problem solving, general skills training, and patient's fears of quitting.
provision of intra-treatment support. The important
• Encourage the patient to talk about the quitting
elements in the support aspect of successful treatment
process. Talk to the patient about the reasons
programs are shown in Figure 5-2-69,10. The common
he/she wants to quit, difficulty encountered while
subjects covered in successful problem solving/skills
quitting, success the patient has achieved, and con-
training programs include: cerns and worries about quitting.
• Recognition of danger signals likely to be associated • Provide basic information about smoking, the
with the risk of relapse, such as being around other risks of continuing, the benefits of quitting, and
smokers, being under time pressure, getting into an the techniques that optimize success. Outline the
argument, drinking alcohol, and negative moods. nature, symptoms, and time course of withdrawal
and techniques for dealing with withdrawal.
• Enhancement of skills needed to handle these
situations, such as learning to anticipate and avoid a
particular stress. psychosocial interventions. Medical contraindications to
nicotine replacement therapy include unstable coronary
• Basic information about smoking and successful artery disease, untreated peptic ulcer disease, and recent
quitting, such as the nature and time course of myocardial infarction or stroke9. Specific studies to date
withdrawal, the addictive nature of smoking, and the do not support the use of nicotine replacement therapy
fact that any return to smoking, including even a single for longer than 8 weeks21, although some patients may
puff, increases the likelihood of a relapse. require extended use to prevent relapse.

Pharmacotherapy. Numerous effective pharmacotherapies All forms of nicotine replacement therapy are significantly
for smoking cessation now exist9-11 (Evidence A), and more effective than placebo. Every effort should be
pharmacotherapy is recommended when counseling is not made to tailor the choice of replacement therapy to the
sufficient to help patients quit smoking. Special consideration individual's culture and lifestyle to improve adherence.
should be given before using pharmacotherapy in selected The patch is generally favored over the gum because it
populations: people with medical contraindications, light requires less training for effective use and is associated
smokers (fewer than 10 cigarettes/day), and pregnant with fewer compliance problems.
and adolescent smokers.
No data are available to help clinicians tailor nicotine
Nicotine replacement products. Numerous studies indicate patch regimens to the intensity of cigarette smoking. In
that nicotine replacement therapy in any form (nicotine all cases it seems generally appropriate to start with the
gum, inhaler, nasal spray, transdermal patch, sublingual higher dose patch. For most patches, which come in
tablet, or lozenge) reliably increases long-term smoking three different doses, patients should use the highest
abstinence rates10,19. Nicotine replacement therapy is dose for the first four weeks and drop to progressively
more effective when combined with counseling and lower doses over an eight-week period. Where only two
behavior therapy20, although nicotine patch or nicotine doses are available, the higher dose should be used for
gum consistently increases smoking cessation rates the first four weeks and the lower dose for the second
regardless of the level of additional behavioral or four weeks.

MANAGEMENT OF COPD 61
GOLD_WR_05 8/18/05 12:56 PM Page 62

When using nicotine gum, the patient needs to be advised The main emphasis should be on primary prevention,
that absorption occurs through the buccal mucosa. For which is best achieved by the elimination or reduction of
this reason, the patient should be advised to chew the exposures to various substances in the workplace.
Secondary prevention, achieved through surveillance and
gum for a while and then put the gum against the inside
early case detection, is also of great importance. Both
of the cheek to allow absorption to occur and prolong
approaches are necessary to improve the present situation
the release of nicotine. Continuous chewing produces
and to reduce the burden of lung disease.
secretions that are swallowed, results in little absorption,
and can cause nausea. Acidic beverages, particularly
coffee, juices, and soft drinks, interfere with the absorption INDOOR AND OUTDOOR
of nicotine. Thus, the patient needs to be advised that
eating or drinking anything except water should be avoided
AIR POLLUTION
for 15 minutes before and during chewing. Although
nicotine gum is an effective smoking cessation treatment, Individuals experience diverse indoor and outdoor
problems with compliance, ease of use, social acceptability, environments throughout the day, each of which has its
risk of developing temporo-mandibular joint symptoms, own unique set of air contaminants. Although outdoor
and unpleasant taste have been noted. In highly dependent and indoor air pollution are generally thought of separately,
smokers, the 4 mg gum is more effective than the 2 mg the concept of total personal exposure may be more
gum22. relevant for COPD. Reducing the risk from indoor and
outdoor air pollution requires a combination of public
Other pharmacotherapy. The antidepressants bupropion23 policy and protective steps taken by individual patients.
and nortriptyline have also been shown to increase long-
term quit rates9,19,24. Although more studies need to be Regulation of Air Quality
conducted with these medications, a randomized
controlled trial with counseling and support showed quit At the national level, achieving a set level of air quality
rates at one year of 30% with sustained-release bupropion should be a high priority; this goal will normally require
alone and 35% with sustained-release bupropion plus legislative action. Details on setting and maintaining air
nicotine patch23. The effectiveness of the antihypertensive quality goals are beyond the scope of this document.
drug clonidine is limited by side effects19.
Understanding health risks posed by local air pollution
OCCUPATIONAL EXPOSURES sources may be difficult and requires skills in community
health, toxicology, and epidemiology. Local physicians may
Although it is not known how many individuals are at risk become involved through concerns about the health of their
of developing respiratory disease from occupational patients or as advocates for the community's environment.
exposures in either developing or developed countries,
many occupationally induced respiratory disorders can be Patient-Oriented Control
reduced or controlled through a variety of strategies aimed
at reducing the burden of inhaled particles and gases25: The health care provider should consider susceptibility
(including family history and exposure to indoor/outdoor
● Implement and enforce strict, legally mandated control pollution) for each individual patient.
of airborne exposure in the workplace.
● Patients should be counseled concerning the nature
● Initiate intensive and continuing education of exposed and degree of their susceptibility. Those who are at
workers, industrial managers, health care workers, high risk should avoid vigorous exercise outdoors
primary care physicians, and legislators. during pollution episodes.
● Educate workers and policymakers on how cigarette ● If various solid fuels are used for cooking and heating,
smoking aggravates occupational lung diseases and adequate ventilation should be encouraged.
why efforts to reduce smoking where a hazard exists
are important. ● Persons with severe COPD should monitor public
announcements of air quality and should stay indoors
when air quality is poor.

62 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 63

● The use of medication should follow the usual clinical 10. The Tobacco Use and Dependence Clinical Practice
indications; therapeutic regimes should not be adjusted Guideline Panel, Staff, and Consortium Representatives.
because of the occurrence of a pollution episode A clinical practice guideline for treating tobacco use and
without evidence of worsening of symptoms or function. dependence. JAMA 2000; 283:244-54.
11. American Medical Association. Guidelines for the diagno-
● Respiratory protective equipment has been developed sis and treatment of nicotine dependence: how to help
for use in the workplace in order to minimize exposure patients stop smoking. Washington, DC: American Medical
to toxic gases and particles. However, under most Association; 1994.
circumstances, health care providers should not suggest 12. Glynn TJ, Manley MW. How to help your patients stop
respiratory protection as a method for reducing the smoking. A National Cancer Institute manual for physi-
risks of ambient air pollution. cians. Bethesda, MD: US Department of Health and
Human Services, Public Health Service, National Institutes
● Air cleaners have not been shown to have health of Health, National Cancer Institute; 1990. NIH Publication
benefits, whether directed at pollutants generated by No. 90-3064.
indoor sources or at those brought in with outdoor air. 13. Glynn TJ, Manley MW, Pechacek TF. Physician-initiated
smoking cessation program: the National Cancer Institute
trials. Prog Clin Biol Res 1990; 339:11-25.
REFERENCES 14. Baillie AJ, Mattick RP, Hall W, Webster P. Meta-analytic
review of the efficacy of smoking cessation interventions.
1. Samet J, Utell MJ. Ambient air pollution. In: Rosenstock L, Drug and Alcohol Review 1994; 13:157-70.
Cullen M, eds. Textbook of occupational and environmental 15. Wilson DH, Wakefield MA, Steven ID, Rohrsheim RA,
medicine. Philadelphia: WB Saunders; 1994. p. 53-60. Esterman AJ, Graham NM. "Sick of smoking": evaluation of
2. Jeffery PK. Cigarette smoke-induced damage of airway a targeted minimal smoking cessation intervention in gen-
mucosa. In: Chretien J, Dusser D, eds. Environmental eral practice. Med J Aust 1990; 152:518-21.
impact on the airways: from injury to repair. Lung biology 16. Britton J, Knox A. Helping people to stop smoking: the new
in health and disease. Vol. 93. New York: Marcel Dekker; smoking cessation guidelines [editorial]. Thorax 1999;
1996. p. 299-354. 54:1-2.
3. Helms PJ. Lung growth: implications for the development of 17. Kottke TE, Battista RN, DeFriese GH, Brekke ML.
disease [editorial]. Thorax 1994; 49:440-1. Attributes of successful smoking cessation interventions in
4. Colley JR, Holland WW, Corkhill RT. Influence of passive medical practice. A meta-analysis of 39 controlled trials.
smoking and parental phlegm on pneumonia and bronchi- JAMA 1988; 259:2883-9.
tis in early childhood. Lancet 1974; 2:1031-4. 18. Ockene JK, Kristeller J, Goldberg R, Amick TL, Pekow PS,
5. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey Hosmer D, et al. Increasing the efficacy of physician-deliv-
WC, Buist AS, et al. Effects of smoking intervention and ered smoking interventions: a randomized clinical trial. J
the use of an inhaled anticholinergic bronchodilator on the Gen Intern Med 1991; 6:1-8.
rate of decline of FEV1. The Lung Health Study. JAMA 19. Lancaster T, Stead L, Silagy C, Sowden A. Effectiveness of
1994; 272:1497-505. interventions to help people stop smoking: findings from
6. World Health Organization. Tobacco free initiative: the Cochrane Library. BMJ 2000; 321:355-8.
policies for public health. Geneva: World Health 20. Schwartz JL. Review and evaluation of smoking cessation
Organization; 1999. Available from: URL: methods: United States and Canada, 1978-1985.
www.who/int/toh/worldnottobacco99 Bethesda, MD: National Institutes of Health; 1987. NIH
7. Tengs TO, Adams ME, Pliskin JS, Safran DG, Siegel JE, Publication No. 87-2940.
Weinstein MC, et al. Five-hundred life-saving interventions 21. Fiore MC, Smith SS, Jorenby DE, Baker TB. The effective-
and their cost-effectiveness. Risk Anal 1995; 15:369-90. ness of the nicotine patch for smoking cessation. A meta-
8. Parrott S, Godfrey C, Raw M, West R, McNeill A. Guidance analysis. JAMA 1994; 271:1940-7.
for commissioners on the cost effectiveness of smoking 22. Sachs DP, Benowitz NL. Individualizing medical treatment
cessation interventions. Health Educational Authority. for tobacco dependence [editorial; comment]. Eur Respir J
Thorax 1998; 53 (Suppl 5 Pt 2): S1-38. 1996; 9:629-31.
9. Fiore MC, Bailey WC, Cohen SJ. Smoking cessation: infor- 23. Tashkin D, Kanner R, Bailey W, Buist S, Anderson P, Nides
mation for specialists. Rockville, MD: US Department of M, Gonzales D, Dozier G, Patel MK, Jamerson B. Smoking
Health and Human Services, Public Health Service, cessation in patients with chronic obstructive pulmonary
Agency for Health Care Policy and Research and Centers disease: a double-blind, placebo-controlled, randomised
for Disease Control and Prevention; 1996. AHCPR trial. Lancet 2001: 19;357 (9268): 1571-5
Publication No. 96-0694.

MANAGEMENT OF COPD 63
GOLD_WR_05 8/18/05 12:56 PM Page 64

24. Jorenby DE, Leischow SJ, Nides MA, Rennard SI,


Johnston JA, Hughes AR, et al. A controlled trial of sus-
tained-release bupropion, a nicotine patch, or both for
smoking cessation. N Engl J Med 1999; 340:685-91.
25. The COPD Guidelines Group of the Standards of Care
Committee of the BTS. BTS guidelines for the manage-
ment of chronic obstructive pulmonary disease. Thorax
1997; 52 Suppl 5:S1-28.

64 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 65

COMPONENT 3: MANAGE STABLE COPD


INTRODUCTION
KEY POINTS:

• The overall approach to managing stable COPD The overall approach to managing stable COPD should
should be characterized by a stepwise increase in be characterized by a stepwise increase in treatment,
treatment, depending on the severity of the disease. depending on the severity of the disease. The step-down
approach used in the chronic treatment of asthma is not
• For patients with COPD, health education can play applicable to COPD since COPD is usually stable and
a role in improving skills, ability to cope with illness, very often progressive. Management of COPD involves
and health status. It is effective in accomplishing several objectives (see Chapter 5, Introduction) that
certain goals, including smoking cessation should be met with minimal side effects from treatment.
(Evidence A). It is based on an individualized assessment of disease
severity (Figure 5-3-1) and response to various therapies.
• None of the existing medications for COPD have
been shown to modify the long-term decline in lung Figure 5-3-1. Factors Affecting the Severity of COPD
function that is the hallmark of this disease
(Evidence A). Therefore, pharmacotherapy for
COPD is used to decrease symptoms and/or • Severity of symptoms
complications. • Severity of airflow limitation
• Frequency and severity of exacerbations
• Bronchodilator medications are central to the • Presence of one or more complications
symptomatic management of COPD (Evidence A). • Presence of respiratory failure
They are given on an as-needed basis or on a • Presence of comorbid conditions
regular basis to prevent or reduce symptoms.
• General health status
• The principal bronchodilator treatments are • Number of medications needed to manage the disease
ß2-agonists, anticholinergics, theophylline, and a
combination of these drugs (Evidence A).
The classification of severity (Figure 1-2) of stable COPD
• Regular treatment with long-acting bronchodilators incorporates an individualized assessment of disease
is more effective and convenient than treatment with severity and therapeutic response into the management
short-acting bronchodilators, but more expensive strategy. This classification is a guide that should help
(Evidence A). health care workers make decisions about the management
of COPD in individual patients. Treatment depends on
• The addition of regular treatment with inhaled the patient’s educational level and willingness to apply
glucocorticosteroids to bronchodilator treatment is the recommended management, on cultural and local
appropriate for symptomatic COPD patients with an conditions, and on the availability of medications.
FEV1 < 50% predicted (Stage III: Severe COPD
and Stage IV: Very Severe COPD) and repeated
exacerbations (Evidence A). EDUCATION
• Chronic treatment with systemic glucocorticosteroids Although patient education is generally regarded as an
should be avoided because of an unfavorable essential component of care for any chronic disease, the
benefit-to-risk ratio (Evidence A). role of education in COPD has been poorly studied.
Assessment of the value of education in COPD may be
• All COPD patients benefit from exercise training difficult because of the relatively long time required to
programs, improving with respect to both exercise achieve improvements in objective measurements of lung
tolerance and symptoms of dyspnea and fatigue function.
(Evidence A).
Studies that have been done indicate that patient education
• The long-term administration of oxygen (> 15 hours alone does not improve exercise performance or lung
per day) to patients with chronic respiratory failure function1-4 (Evidence B), but it can play a role in improving
has been shown to increase survival (Evidence A).
skills, ability to cope with illness, and health status5.

MANAGEMENT OF COPD 65
GOLD_WR_05 8/18/05 12:56 PM Page 66

These outcomes are not traditionally measured in clinical Components of an Education Program
trials, but they may be most important in COPD where
even pharmacologic interventions generally confer only a The topics that seem most appropriate for an education
small benefit in terms of lung function. program include: smoking cessation; basic information
about COPD and pathophysiology of the disease; general
Patient education regarding smoking cessation has the approach to therapy and specific aspects of medical
greatest capacity to influence the natural history of treatment; self-management skills; strategies to help
COPD. Evaluation of the smoking cessation component minimize dyspnea; advice about when to seek help; self-
in a long-term, multicenter study indicates that if effective management and decision-making during exacerbations;
resources and time are dedicated to smoking cessation, and advance directives and end-of-life issues (Figure 5-3-2).
25% long-term quit rates can be maintained6 (Evidence Education should be part of consultations with health
A). Education also improves patient response to care workers beginning at the time of first assessment for
exacerbations7,8 (Evidence B). Prospective end-of-life
discussions can lead to understanding of advance Figure 5-3-2. Topics for Patient Education
directives and effective therapeutic decisions at the end
of life9 (Evidence B). Stage 0: At Risk
• Information and advice about reducing risk factors
Ideally, educational messages should be incorporated
into all aspects of care for COPD and may take place Stage I: Mild COPD through Stage III: Severe COPD
in many settings: consultations with physicians or other Above topic, plus:
health care workers, home-care or outreach programs, • Information about the nature of COPD
and comprehensive pulmonary rehabilitation programs. • Instruction on how to use inhalers and other treatments
• Recognition and treatment of exacerbations
Goals and Educational Strategies • Strategies for minimizing dyspnea

It is vital for patients with COPD to understand the nature Stage IV: Very Severe COPD
of their disease, risk factors for progression, and their role Above topics, plus:
and the role of health care workers in achieving optimal • Information about complications
management and health outcomes. Education should be • Information about oxygen treatment
tailored to the needs and environment of the individual • Advance directives and end-of-life decisions
patient, interactive, directed at improving quality of life,
simple to follow, practical, and appropriate to the intellectual COPD and continuing with each follow-up visit. The
and social skills of the patient and the caregivers. intensity and content of these educational messages
should vary depending on the severity of the patient’s
In managing COPD, open communication between patient disease. In practice, a patient often poses a series of
and physician is essential. In addition to being empathic, questions to the physician (Figure 5-3-3). It is important
attentive and communicative, health professionals should to answer these questions fully and clearly, as this may
pay attention to patients’ fears and apprehensions, focus help make treatment more effective.
on educational goals, tailor treatment regimens to each
individual patient, anticipate the effect of functional There are several different types of educational programs,
decline, and optimize patients’ practical skills. ranging from simple distribution of printed materials, to
teaching sessions designed to convey information about
Several specific education strategies have been shown to COPD, to workshops designed to train patients in specific
improve patient adherence to medication and management skills (e.g., self-management, peak flow monitoring).
regimens. In COPD, adherence does not simply refer to
whether patients take their medication appropriately. It Although printed materials may be a useful adjunct to
also covers a range of non-pharmacologic treatments - other educational messages, passive dissemination of
e.g., maintaining an exercise program after pulmonary printed materials alone does not improve skills or health
rehabilitation, undertaking and sustaining smoking outcomes. Education is most effective when it is interactive
cessation, and using devices such as nebulizers, spacers, and conducted in small workshops4 (Evidence B)
designed to improve both knowledge and skills.
and oxygen concentrators properly.
Behavioral approaches such as cognitive therapy and
behavior modification lead to more effective self-
management skills and maintenance of exercise programs.

66 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 67

COPD have been shown to modify the long-term decline


Figure 5-3-3. Examples of Patient Questions in lung function that is the hallmark of this disease6,11-13
• What is COPD? (Evidence A). However, this should not preclude efforts
to use medications to control symptoms. Since COPD
• What causes COPD?
is usually progressive, recommendations for the
• How will it affect me?
pharmacological treatment of COPD reflect the following
• Can it be treated?
general principles:
• What will happen if my disease gets worse?
• What will happen if I need to be admitted to the • There should be a stepwise increase in treatment,
hospital? depending on the severity of the disease. (The
• How will I know when I need oxygen at home? step-down approach used in the chronic treatment
• What if I do not wish to be admitted to intensive of asthma is not applicable to COPD.)
care for ventilation?
Answers to these questions can be developed from this document • Regular treatment needs to be maintained at the same
and will depend on local circumstances. In all cases, it is important level for long periods of time unless significant side
that answers are clear and use terminology that the patient understands. effects occur or the disease worsens.

Cost Effectiveness of Education Programs • Treatment response of an individual patient is variable


for COPD Patients and should be monitored closely and adjusted frequently.

The cost effectiveness of education programs for COPD The medications are presented in the order in which they
patients is highly dependent on local factors that influence would normally be introduced in patient care, based on
the cost of access to medical services and that will vary the level of disease severity. However, each treatment
regimen needs to be patient-specific as the relationship
substantially between countries. In one cost-benefit
between the severity of symptoms and the severity of air-
analysis of education provided to hospital inpatients with
flow limitation is influenced by other factors, such as the
COPD10, an information package resulted in increased
frequency and severity of exacerbations, the presence of
knowledge of COPD and reduced use of health services,
one or more complications, the presence of respiratory
including reductions of hospital readmissions and general
failure, comorbidities (cardiovascular disease, sleep-related
practice consultations. The education package involved
disorders, etc.), and general health status.
training patients to increase knowledge of COPD,
medication usage, precautions for exacerbations, and
peak flow monitoring technique. However, this study
Bronchodilators
was undertaken in a heterogeneous group of patients -
Medications that increase the FEV1 or change other
65% were smokers and 88% were judged to have an
spirometric variables, usually by altering airway smooth
asthmatic component to their disease - and these
muscle tone, are termed bronchodilators14, since the
findings may not hold true for a “pure” COPD population.
improvements in expiratory flow reflect widening of the
In a study of mild to moderate COPD patients at an out-
airways rather than changes in lung elastic recoil. Such
patient clinic, patient education involving one four hour
drugs improve emptying of the lungs, tend to reduce
group session followed by one to two individual nurse- dynamic hyperinflation at rest and during exercise15,
and physiotherapist-sessions improved patient outcomes and improve exercise performance. The extent of these
and reduced costs in a 12-month follow-up220. changes, especially in moderate to severe disease, is
not easily predictable from the improvement in FEV116,17.
Although a healthy lifestyle is important, and should be Regular bronchodilation with drugs that act primarily on
encouraged, additional studies are needed to identify airway smooth muscle does not modify the decline of
specific components of self-management programs that function in mild COPD and, by inference, the prognosis
are effective200. of the disease6 (Evidence B).

PHARMACOLOGIC TREATMENT Bronchodilator medications are central to the symptomatic


management of COPD18-21 (Evidence A). They are given
Overview of the Medications either on an as-needed basis for relief of persistent or
worsening symptoms, or on a regular basis to prevent or
Pharmacologic therapy is used to prevent and control reduce symptoms. The side effects of bronchodilator
symptoms, reduce the frequency and severity of therapy are pharmacologically predictable and dose
exacerbations, improve health status, and improve dependent. Adverse effects are less likely, and resolve
exercise tolerance. None of the existing medications for more rapidly after treatment withdrawal, with inhaled than

MANAGEMENT OF COPD 67
GOLD_WR_05 8/18/05 12:56 PM Page 68

with oral treatment. However, COPD patients tend to be cholinergic by an order of magnitude, especially when
older than asthma patients and more likely to have given by a wet nebulizer, appears to provide subjective
comorbidities, so their risk of developing side effects is benefit in acute episodes25 (Evidence B) but is not
greater. necessarily helpful in stable disease26 (Evidence C).

When treatment is given by the inhaled route, attention to Inhaled ß2-agonists have a relatively rapid onset of
effective drug delivery and training in inhaler technique is bronchodilator effect although this is probably slower in
essential. COPD patients may have more problems in COPD than in asthma. The bronchodilator effects of
effective coordination and find it harder to use a simple short-acting ß2-agonists usually wear off within 4 to 6
Metered Dose Inhaler (MDI) than do healthy volunteers hours27,28 (Evidence A). For single-dose, as needed use
or younger asthmatics. It is essential to ensure that inhaler in COPD, there appears to be no advantage in using leval-
technique is correct and to re-check this at each visit. buterol over conventional nebulized bronchodilators201.
Long-acting inhaled ß2-agonists, such as salmeterol and
Alternative breath-activated or spacer devices are available formoterol, show a duration of effect of 12 hours or more
for most formulations. Dry powder inhalers may be more with no loss of effectiveness overnight or with regular use
convenient and possibly provide improved drug deposition, in COPD patients29-32 (Evidence A). The long-acting
although this has not been established in COPD. In inhaled anticholinergic, tiotropium, has a duration of
general, particle deposition will tend to be more central action of more than 24 hours (Evidence A)33-35.
with the fixed airflow limitation and lower inspiratory flow
rates in COPD22,23. All categories of bronchodilators have been shown to
increase exercise capacity in COPD, without necessarily
Wet nebulizers are not recommended for regular treatment producing significant changes in FEV136,37,221,222 (Evidence A).
because they are more expensive and require appropriate Regular treatment with long-acting bronchodilators is
maintenance24. A list of currently available inhaler devices more effective and convenient than treatment with
is provided at http://www.goldcopd.org/inhalers/. The short-acting bronchodilators, but more expensive34,38,39,223
choice will depend on availability, cost, the prescribing (Evidence A).
physician, and the skills and ability of the patient.

Dose-response relationships using the FEV1 as the


outcome are relatively flat with all classes of
bronchodilators18-21. The dose-response relationships for Figure 5-3-5. Dose-Response
short-acting anticholinergics and ß2-agonists are shown Relationships for Short-acting Bronchodilators21
in Figure 5-3-521. Toxicity is also dose related.
Increasing the dose of either a ß2-agonist or an anti-

Figure 5-3-4. Bronchodilators in Stable COPD

• Bronchodilator medications are central to symptom


management in COPD.

• Inhaled therapy is preferred.

• The choice between ß2-agonist, anticholinergic,


theophylline, or combination therapy depends on
availability and individual response in terms of
symptom relief and side effects.

• Bronchodilators are prescribed on an as-needed or Cumulative dose (µg)


on a regular basis to prevent or reduce symptoms.
Open symbols - salbutamol (ß2-agonist)
• Long-acting inhaled bronchodilators are more
Closed symbols - ipratropium (anticholinergic)
effective and convenient, but more expensive.
Squares - patients with asthma
• Combining bronchodilators may improve efficacy Triangles - patients with COPD
and decrease the risk of side effects compared to
increasing the dose of a single bronchodilator. Printed with permission from Higgins BG, Powell RM, Cooper S, Tattersfield AE. European
Respiratory Journal 1991; 4:415-20. Copyright 1991 European Respiratory Society Journals, Ltd.

68 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 69

Figure 5-3-6. Commonly Used Formulations of Drugs used in COPD


Drug Inhaler Solution for Oral Vials for Injection Duration of Action
(g) Nebulizer (mg/ml) (mg) (hours)

2-agonists
Short-acting
Fenoterol 100-200 (MDI) 1 0.05% (Syrup) 4-6
Salbutamol (albuterol) 100, 200 (MDI & DPI) 5 5mg (Pill) 0.1, 0.5 4-6
Syrup 0.024%
Terbutaline 400, 500 (DPI) – 2.5, 5 (Pill) 0.2, 0.25 4-6

Long-acting
Formoterol 4.5–12 (MDI & DPI) 12+
Salmeterol 25-50 (MDI & DPI) 12+

Anticholinergics
Short-acting
Ipratropium bromide 20, 40 (MDI) 0.25-0.5 6-8
Oxitropium bromide 100 (MDI) 1.5 7-9

Long-acting
Tiotropium 18 (DPI) 24+

Combination short-acting 2-agonists plus anticholinergic in one inhaler


Fenoterol/Ipratropium 200/80 (MDI) 1.25/0.5 6-8
Salbutamol/Ipratropium 75/15 (MDI) 0.75/4.5 6-8

Methylxanthines
Aminophylline 200-600 mg (Pill) 240 mg Variable, up to 24
Theophylline (SR) 100-600 mg (Pill) Variable, up to 24

Inhaled glucocorticosteroids
Beclomethasone 50-400 (MDI & DPI) 0.2-0.4
Budesonide 100, 200, 400 (DPI) 0.20, 0.25, 0.5
Fluticasone 50-500 (MDI & DPI)
Triamcinolone 100 (MDI) 40 40

Combination long-acting 2-agonists plus glucocorticosteroids in one inhaler


Formoterol/Budesonide 4.5/80, 160 (DPI)
(9/320) (DPI)
Salmeterol/Fluticasone 50/100, 250, 500 (DPI)
25/50, 125, 250 (MDI)
Systemic glucocorticosteroids
Prednisone 5-60 mg (Pill)
Methyl-prednisolone 10-2000 mg 4, 8, 16 mg (Pill)

MDI=metered dose inhaler; DPI=dry powder inhaler

Regular use of a long-acting ß2-agonist38 or a short- or COPD, ß2-agonists, anticholinergics, and methylxanthines,
long-acting anticholinergic improves health status34,38,39. are shown in Figure 5-3-6. The choice depends on the
Theophylline is effective in COPD, but due to its potential availability of medication and the patient’s response.
toxicity inhaled bronchodilators are preferred when available.
All studies that have shown efficacy of theophylline in ß2-agonists. The principal action of ß2-agonists is to
COPD were done with slow-release preparations. The relax airway smooth muscle by stimulating ß2-adrenergic
classes of bronchodilator drugs commonly used in treating receptors, which increases cyclic AMP and produces

MANAGEMENT OF COPD 69
GOLD_WR_05 8/18/05 12:56 PM Page 70

functional antagonism to bronchoconstriction. Oral Use of wet nebulizer solutions with a face mask has been
therapy is slower in onset and has more side effects than reported to precipitate acute glaucoma, probably by a
inhaled treatment40 (Evidence A). direct effect of the solution on the eye. Mucociliary
clearance is unaffected by these drugs, and respiratory
Adverse effects. Stimulation of ß2-receptors can produce infection rates are not increased.
resting sinus tachycardia and has the potential to precipitate
cardiac rhythm disturbances in very susceptible patients, Methylxanthines. Controversy remains about the exact
although this appears to be a remarkably rare event with effects of xanthine derivatives. They may act as non-
inhaled therapy. Exaggerated somatic tremor is trouble- selective phosphodiesterase inhibitors, but have also
some in some older patients treated with higher doses of been reported to have a range of non-bronchodilator
ß2-agonists, whatever the route of administration, and actions, the significance of which is disputed47-51. Data on
this limits the dose that can be tolerated. Although duration of action for conventional, or even slow-release,
hypokalemia can occur, especially when treatment is xanthine preparations are lacking in COPD. Changes in
combined with thiazide diuretics41, and oxygen consumption inspiratory muscle function have been reported in
can be increased under resting conditions42, these metabolic patients treated with theophylline47, but whether this
effects show tachyphylaxis unlike the bronchodilator actions. reflects changes in dynamic lung volumes or a primary
Mild falls in PaO2 occur after administration of both short- effect on the muscle is not clear (Evidence B). All studies
and long-acting ß2-agonists43, but the clinical significance that have shown efficacy of theophylline in COPD were
of these changes is doubtful. Despite the concerns done with slow-release preparations. Theophylline is
raised some years ago, further detailed study has found effective in COPD but, due to its potential toxicity, inhaled
no association between ß2-agonist use and an accelerated bronchodilators are preferred when available.
loss of lung function or increased mortality in COPD.
Adverse effects. Toxicity is dose related, a particular
Anticholinergics. The most important effect of problem with the xanthine derivatives because their
anticholinergic medications in COPD patients appears therapeutic ratio is small and most of the benefit occurs
to be blockage of acetylcholine’s effect on M3 receptors. only when near-toxic doses are given49,50 (Evidence A).
Current short-acting drugs also block M2 receptors and
Methylxanthines are nonspecific inhibitors of all phospho-
modify transmission at the pre-ganglionic junction,
diesterase enzyme subsets, which explains their
although these effects appear less important in COPD44.
The long-acting tiotropium has a pharmacokinetic
selectivity for the M3 and the M1 receptor45.

The bronchodilating effect of short-acting inhaled


anticholinergics lasts longer than that of short-acting
ß2-agonists, with some bronchodilator effect generally
Figure 5-3-7. Drugs and Physiological Variables
apparent up to 8 hours after administration27 (Evidence
that Affect Theophylline Metabolism in COPD
A). The long-acting inhaled anticholinergic, tiotropium,
has a duration of action of more than 24 hours33-35
(Evidence A). Increased Decreased
• Tobacco smoking • Old age
Adverse effects. Anticholinergic drugs, such as ipratropium, • Anticonvulsant drugs • Arterial hypoxemia
oxitropium and tiotropium bromide, are poorly absorbed • Rifampicin (PaO2 < 6.0 kPa, 45
which limits the otherwise troublesome systemic effects • Alcohol mm Hg)
seen with atropine. Extensive use of this class of inhaled • Respiratory acidosis
agents in a wide range of doses and clinical settings has • Congestive cardiac
shown them to be very safe. The main side effect is dryness failure
of the mouth. Twenty-one days of inhaled tiotropium, 18 • Liver cirrhosis
µg day as a dry powder, does not retard mucus clearance • Erythromycin
from the lungs224. Although occasional prostatic symptoms • Quinolone antibiotics
have been reported, there are no data to prove a true • Cimetidine
causal relationship. A bitter, metallic taste is reported (not ranitidine)
by some patients using ipratropium. An unexpected • Viral infections
small increase in cardiovascular events in COPD patients
• Herbal remedies
regularly treated with ipratropium bromide has been
(St. John’s Wort)
reported and requires further investigation46.

70 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 71

wide range of toxic effects. Problems include the but do show significant bronchodilation after a short
development of atrial and ventricular arrhythmias (which course of oral glucocorticosteroids.
can prove fatal) and grand mal convulsions (which can
occur irrespective of prior epileptic history). More There is mounting evidence, however, that a short course
common and less dramatic side effects include of oral glucocorticosteroids is a poor predictor of the long-
headaches, insomnia, nausea, and heartburn, and term response to inhaled glucocorticosteroids in COPD13,59.
these may occur within the therapeutic range of serum For this reason, there appears to be insuffi-cient evidence
theophylline. Unlike the other bronchodilator classes, to recommend a therapeutic trial with oralglucocorticos-
xanthine derivatives may involve a risk of overdose teroids in patients with Stage II: Moderate COPD, Stage
(either intentional or accidental). III: Severe COPD, or Stage IV: Very Severe COPD and
poor response to an inhaled bronchodilator.
Theophylline, the most commonly used methylxanthine,
is metabolized by cytochrome P450 mixed function Oral glucocorticosteroids - long-term. Two retrospective
oxidases. Clearance of the drug declines with age. studies60,61 analyzed the effects of treatment with oral
Many other physiological variables and drugs modify glucocorticosteroids on long-term FEV1 changes in clinic
theophylline metabolism; some of the potentially populations of patients with moderate to very severe
important interactions are listed in Figure 5-3-7. COPD. The retrospective nature of these studies, the lack
of a true control group, and the imprecise definition of
Combination bronchodilator therapy. Combining COPD are reasons for a cautious interpretation of the
bronchodilators with different mechanisms and durations data and conclusions.
of action may increase the degree of bronchodilation for
equivalent or lesser side effects. A combination of a A side effect of long-term treatment with systemic
short-acting ß2-agonist and an anticholinergic produces glucocorticosteroids is steroid myopathy62-64, which
greater and more sustained improvements in FEV1 contributes to muscle weakness, decreased functionality,
than either drug alone and does not produce evidence and respiratory failure in subjects with advanced COPD.
of tachyphylaxis over 90 days of treatment27,52,53 In view of the well-known toxicity of long-term treatment
(Evidence A). with oral glucocorticosteroids, it is not surprising that no
prospective studies have been performed on the long-
The combination of a ß2-agonist, an anticholinergic, term effects of these drugs in COPD.
and/or theophylline may produce additional improvements
in lung function27,51,54-56 and health status27,57. Increasing Therefore, based on the lack of evidence of benefit, and
the number of drugs usually increases costs, and an the large body of evidence on side effects, long-term
equivalent benefit may occur by increasing the dose of treatment with oral glucocorticosteroids is not recommended
one bronchodilator when side effects are not a limiting in COPD (Evidence A).
factor. Detailed assessments of this approach have not
been carried out. Inhaled glucocorticosteroids. Regular treatment with
inhaled glucocorticosteroids does not modify the long-
Glucocorticosteroids term decline of FEV1 in patients with COPD11-13,65.
However, regular treatment with inhaled glucocorti-
The effects of oral and inhaled glucocorticosteroids in costeroids is appropriate for symptomatic COPD patients
COPD are much less dramatic than in asthma, and their with an FEV1 < 50% predicted (Stage III: Severe COPD
role in the management of stable COPD is limited to very and Stage IV: Very Severe COPD) and repeated
specific indications. The use of glucocorticosteroids for exacerbations (for example, 3 in the last three years)66-69
the treatment of acute exacerbations is described in (Evidence A). This treatment has been shown to reduce
Component 4: Manage Exacerbations. the frequency of exacerbations and thus improve health
status224 (Evidence A), and withdrawal from treatment
Oral glucocorticosteroids - short-term. Many existing with inhaled glucocorticosteroids can lead to exacerbations
COPD guidelines recommend the use of a short course in some patients202. Inhaled glucocorticosteroid combined
(two weeks) of oral glucocorticosteroids to identify COPD with a long-acting ß2-agonist is more effective than the
patients who might benefit from long-term treatment with individual components66,68,69,203,204 (Evidence A). Short-
oral or inhaled glucocorticosteroids. This recommendation term treatment with a combined inhaled glucocorticosteroid
is based on evidence58 that short-term effects predict and long-acting ß2-agonist resulted in greater control of
long-term effects of oral glucocorticosteroids on FEV1, lung function and symptoms than combined anticholinergic
and evidence that asthma patients with airflow limitation and short-acting ß2-agonist225.
might not respond acutely to an inhaled bronchodilator

MANAGEMENT OF COPD 71
GOLD_WR_05 8/18/05 12:56 PM Page 72

Figure 5-3-8. Therapy at Each Stage of COPD


Old 0: At Risk I: Mild II: Moderate III: Severe
IIA IIB

New 0: At Risk I: Mild II: Moderate III: Severe IV: Very Severe

Characteristics • Chronic symptoms • FEV1/FVC < 70% • FEV1/FVC < 70% • FEV1/FVC < 70% • FEV1/FVC < 70%
• Exposure to risk • FEV1 ≥ 80% • 50% ≤ FEV1 < 80% • 30% ≤ FEV1 < 50% • FEV1 < 30% or FEV1 < 50%
factors • With or without • With or without • With or without predicted plus chronic
• Normal spirometry symptoms symptoms symptoms respiratory failure

Avoidance of risk factor(s); influenza vaccination


Add short-acting bronchodilator when needed
Add regular treatment with one or more
long-acting bronchodilators
Add rehabilitation
Add inhaled glucocorticosteroids
if repeated exacerbations

Add long-term
oxygen if chronic
respiratory
failure
Consider surgical
treatments

The dose-response relationships and long-term safety Pharmacologic Therapy by Disease Severity
of inhaled glucocorticosteroids in COPD are not known.
Only moderate to high doses have been used in long- term Figure 5-3-8 provides a summary of recommended
clinical trials. Two studies showed an increased incidence treatment at each stage of COPD. For patients with few
of skin bruising in a small percentage of the COPD or intermittent symptoms (Stage I: Mild COPD), short-
patients11,13. One long-term study showed no effect of acting inhaled therapy as needed to control dyspnea or
budesonide on bone density and fracture rate11,70, while coughing spasms is sufficient. If inhaled bronchodilators
another study showed that treatment with triamcinolone are not available, regular treatment with slow-release
acetonide was associated with a decrease in bone theophylline should be considered.
density65. The efficacy and side effects of inhaled
glucocorticosteroids in asthma are dependent on the dose In patients with Stage II: Moderate COPD to Stage IV:
and type of glucocorticosteroid71. This pattern can also Very Severe COPD) whose symptoms are not adequately
be expected in COPD and needs documentation in this controlled with as-needed short-acting bronchodilators,
patient population. Treatment with inhaled glucocortico- adding regular treatment with a long-acting inhaled
steroids can be recommended for patients with more bronchodilator is recommended (Evidence A). Regular
advanced COPD and repeated exacerbations. treatment with long-acting bronchodilators is more effective
and convenient than treatment with short-acting

72 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 73

bronchodilators, but more expensive (Evidence A). There Antibiotics. In several large-scale controlled studies78-80,
is insufficient evidence to favor one or the other long- prophylactic, continuous use of antibiotics was shown
acting bronchodilator. For patients who need additional to have no effect on the frequency of exacerbations in
symptom control, adding theophylline leads to additional COPD. Another study examined the efficacy of winter
benefits. chemoprophylaxis over a period of 5 years and concluded
that there was no benefit81. Thus, on the present evidence,
Patients with Stage II: Moderate COPD to Stage IV: the use of antibiotics, other than for treating infectious
Very Severe COPD who are on regular short- or long- exacerbations of COPD and other bacterial infections, is
acting bronchodilator therapy may also use a short-acting not recommended82,83 (Evidence A).
bronchodilator as needed.
Mucolytic (mucokinetic, mucoregulator) agents
Some patients may request regular treatment with (ambroxol, erdosteine, carbocysteine, iodinated glycerol).
high-dose nebulized bronchodilators, especially if they The regular use of mucolytics in COPD has been
have experienced subjective benefit from this treatment evaluated in a number of long-term studies with
during an acute exacerbation. Clear scientific evidence controversial results84-86. The majority showed no effect
for this approach is lacking, but one suggested option is of mucolytics on lung function or symptoms, although
to examine the improvement in mean daily peak some have reported a reduction in the frequency of
expiratory flow recording during two weeks of treatment exacerbations. A Cochrane collaborative review
in the home and continue with nebulizer therapy if a performed a meta-analysis of all the available data,
significant change occurs24. In general, nebulized therapy including that from a number of abstracts87. A statistically
for a stable patient is not appropriate unless it has been significant reduction in the number of episodes of chronic
shown to be better than conventional dose therapy. bronchitis was found in patients treated with mucolytics
compared to those receiving placebo. However, these
In patients with a postbronchodilator FEV1 < 50% data are not easy to interpret, as the follow-up ranged
predicted (Stage III: Severe COPD to Stage IV: Very from 2 to 6 months and the patients all had an FEV1 >
Severe COPD) and a history of repeated exacerbations 50% predicted. Although a few patients with viscous
(for example, three in the last three years), regular sputum may benefit from mucolytics88,89, the overall
treatment with inhaled glucocorticosteroids reduce benefits seem to be very small. Therefore, the wide-
frequency of exacerbations and improve health status. spread use of these agents cannot be recommended
In these patients, regular treatment with an inhaled on the basis of the present evidence (Evidence D).
glucocorticosteroid should be added to regular
bronchodilator treatment. Chronic treatment with oral Antioxidant agents. Antioxidants, in particular
glucocorticosteroids should be avoided. N-acetylcysteine, have been shown to reduce the
frequency of exacerbations and could have a role in the
Other Pharmacologic Treatments treatment of patients with recurrent exacerbations90-93
(Evidence B). However, before their routine use can be
Vaccines. Influenza vaccines can reduce serious recommended, the results of ongoing trials will have to
illness226 and death in COPD patients by about 50%72, 205 be carefully evaluated.
(Evidence A). Vaccines containing killed or live, inactivated
viruses are recommended73 as they are more effective in Immunoregulators (immunostimulators,
elderly patients with COPD74. The strains are adjusted immunomodulators). Studies using an immunostimulator
each year for appropriate effectiveness and should be in COPD show a decrease in the severity and frequency
given once (in autumn) or twice (in autumn and winter) of exacerbations94,227. However, additional studies to
each year. A pneumococcal vaccine containing 23 examine the long term effects of this therapy are required
virulent serotypes has been used, but sufficient data to before regular use can be recommended95 (Evidence B).
support its general use in COPD patients are lacking75-77
(Evidence B). Antitussives. Cough, although sometimes a troublesome
symptom in COPD, has a significant protective role96.
Alpha-1 antitrypsin augmentation therapy. Young Thus the regular use of antitussives is contraindicated in
patients with severe hereditary alpha-1 antitrypsin stable COPD (Evidence D).
deficiency and established emphysema may be
candidates for alpha-1 antitrypsin augmentation therapy.
Vasodilators. The belief that pulmonary hypertension
However, this therapy is very expensive, is not available
in COPD is associated with a poorer prognosis has
in most countries, and is not recommended for patients
provoked many attempts to reduce right ventricular
with COPD that is unrelated to alpha-1 antitrypsin
afterload, increase cardiac output, and improve oxygen
deficiency (Evidence C).

MANAGEMENT OF COPD 73
GOLD_WR_05 8/18/05 12:56 PM Page 74

delivery and tissue oxygenation. Many agents have


been evaluated, including inhaled nitric oxide, but the Figure 5-3-9. The Cycle of Physical, Social, and
results have been uniformly disappointing. In patients Psychosocial Consequences of COPD
with COPD, in whom hypoxemia is caused primarily by
venti-lation-perfusion mismatching rather than by Lack of Fitness
increased intrapulmonary shunt (as in noncardiogenic
pulmonary edema), inhaled nitric oxide can worsen gas
exchange because of altered hypoxic regulation of
ventilation-perfusion balance97,98. Therefore, based on
the available evidence, nitric oxide is contraindicated in COPD Dyspnea Immobility
stable COPD.

Respiratory stimulants. Almitrine bismesylate, a relatively


specific peripheral chemoreceptor stimulant that increases Depression Social Isolation
ventilation at any level of CO2 under hypoxemic conditions,
has been studied in both stable respiratory failure and
exacerbations. It improves ventilation-perfusion relation-
ships by modifying the hypoxic vasoconstrictor response. Stage II: Moderate COPD, Stage III: Severe COPD,
Oral almitrine has been shown to improve oxygenation, and Stage IV: Very Severe COPD, include exercise
but to a lesser degree than low doses of inspired O2. deconditioning, relative social isolation, altered mood
There is no evidence that almitrine improves survival or states (especially depression), muscle wasting, and
quality of life, and in large clinical trials it was associated weight loss. These problems have complex interrelation-
with a number of significant side effects, particularly ships and improvement in any one of these interlinked
peripheral neuropathy99-101. Therefore, on the present processes can interrupt the “vicious circle” in COPD so
evidence almitrine is not recommended for regular use in that positive gains occur in all aspects of the illness
stable COPD patients (Evidence B). The use of (Figure 5-3-9).
doxapram, a non-specific but relatively safe respiratory
stimulant available as an intravenous formulation, is not
recommended in stable COPD (Evidence D).

Narcotics (morphine). The use of oral and parenteral


opioids are effective for treating dyspnea in COPD
patients with advanced disease. There are insufficient
Figure 5-3-10. Benefits of Pulmonary Rehabilitation
data to conclude whether nebulized opioids are
in COPD5,110-120
effective102. However, some clinical studies suggest that
morphine used to control dyspnea may have serious
adverse effects and its benefits may be limited to a few • Improves exercise capacity (Evidence A).
sensitive subjects103-107. • Reduces the perceived intensity of breathlessness
(Evidence A).
Others. Nedocromil, leukotriene modifiers, and alternative • Can improve health-related quality of life (Evidence A).
healing methods (e.g., herbal medicine, acupunture, • Reduces the number of hospitalizations and days in
homeopathy) have not been adequately tested in COPD
the hospital (Evidence A).
patients and thus cannot be recommended at this time.
• Reduces anxiety and depression associated with
COPD (Evidence A).
NON-PHARMACOLOGIC TREATMENT • Strength and endurance training of the upper limbs
improves arm function (Evidence B).
Rehabilitation • Benefits extend well beyond the immediate period of
training (Evidence B).
The principal goals of pulmonary rehabilitation are to
• Improves survival (Evidence B).
reduce symptoms, improve quality of life, and increase
physical and emotional participation in everyday activities. • Respiratory muscle training is beneficial, especially
To accomplish these goals, pulmonary rehabilitation when combined with general exercise training
covers a range of non-pulmonary problems that may not (Evidence C).
be adequately addressed by medical therapy for COPD. • Psychosocial intervention is helpful (Evidence C).
Such problems, which especially affect patients with

74 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 75

Pulmonary rehabilitation has been carefully evaluated in Components of pulmonary rehabilitation programs.
a large number of clinical trials; the various benefits are The components of pulmonary rehabilitation vary
summarized in Figure 5-3-105,108-118. widely from program to program but a comprehensive
pulmonary rehabilitation program includes exercise
Patient selection and program design. Although training, nutrition counseling, and education.
more information is needed on criteria for patient
selection for pulmonary rehabilitation programs, COPD Exercise training. Exercise tolerance can be assessed
patients at all stages of disease appear to benefit from by either bicycle ergometry or treadmill exercise with the
exercise training programs, improving with respect to measurement of a number of physiological variables,
both exercise tolerance and symptoms of dyspnea and including maximum oxygen consumption, maximum heart
fatigue119 (Evidence A). Data suggest that these rate, and maximum work performed. A less complex
benefits can be sustained even after a single pulmonary approach is to use a self-paced, timed walking test
rehabilitation program120-122. (e.g., 6-minute walking distance). These tests require at
least one practice session before data can be interpreted.
Benefit does wane after a rehabilitation program ends, Shuttle walking tests offer a compromise: they provide
but if exercise training is maintained at home the patient’s more complete information than an entirely self-paced
health status remains above pre-rehabilitation levels test, but are simpler to perform than a treadmill test125.
(Evidence B). To date there is no consensus on whether
repeated rehabilitation courses enable patients to sustain Exercise training ranges in frequency from daily to
the benefits gained through the initial course. weekly, in duration from 10 minutes to 45 minutes per
session, and in intensity from 50% peak oxygen
Ideally, pulmonary rehabilitation should involve several consumption (VO2 max) to maximum tolerated126. The
types of health professionals. Significant benefits can optimum length for an exercise program has not been
also occur with more limited personnel, as long as investigated in randomized, controlled trials. Thus, the
dedicated professionals are aware of the needs of each length depends on the resources available and usually
patient. Benefits have been reported from rehabilitation
ranges from 4 to 10 weeks, with longer programs
programs conducted in inpatient, outpatient, and home
resulting in larger effects than shorter programs111.
settings112,113,123. Considerations of cost and availability
most often determine the choice of setting. The
Participants are often encouraged to achieve a
educational and exercise training components of
predetermined target heart rate127, but this goal may have
rehabilitation are usually conducted in groups, normally
limitations in COPD. In many programs, especially those
with 6 to 8 individuals per class (Evidence D).
using simple corridor exercise training, the patient is
The following points summarize current knowledge of encouraged to walk to a symptom-limited maximum, rest,
considerations important in choosing patients: and then continue walking until 20 minutes of exercise
have been completed. Use of a simple wheeled walking-
Functional status: Benefits have been seen in patients aid seems to improve walking distance and reduces
with a wide range of disability, although those who are breathlessness in severely disabled COPD patients
chair-bound appear unlikely to respond even to home vis- (Evidence C)206-208. The minimum length of an effective
iting programs124 (Evidence A). rehabilitation program is two months; the longer the
program continues, the more effective the results
Severity of dyspnea: Stratification by breathlessness (Evidence B)128-130. However, as yet, no effective program
intensity using the MRC questionnaire (Figure 5-1-3) has been developed to maintain the effects over time131.
may be helpful in selecting patients most likely to benefit Many physicians advise patients unable to participate in
from rehabilitation. Those with MRC grade 5 dyspnea a structured program to exercise on their own (e.g., walking
may not benefit124 (Evidence B). 20 minutes daily). The benefits of this general advice
have not been tested, but it is reasonable to offer such
Motivation: Selecting highly motivated participants is advice to patients if a formal program is not available.
especially important in the case of outpatient programs121.
Some programs also include upper limb exercises,
Smoking status: There is no evidence that smokers will usually involving an upper limb ergometer or resistive
benefit less than nonsmokers, but many clinicians training with weights. There are no randomized clinical
believe that inclusion of a smoker in a rehabilitation trial data to support the routine inclusion of these
program should be conditional on their participation in a exercises, but they may be helpful in patients with
smoking cessation program. Some data indicate that con- comorbidities that restrict other forms of exercise and
tinuing smokers are less likely to complete pulmonary those with evidence of respiratory muscle weakness132,133.
rehabilitation programs than nonsmokers121 (Evidence B). The addition of upper limb exercises or other strength

MANAGEMENT OF COPD 75
GOLD_WR_05 8/18/05 12:56 PM Page 76

training to aerobic training is effective in improving strength, The first two assessments are important for establishing
but does not improve quality of life or exercise tolerance134. entry suitability and baseline status but are not used in
outcome assessment. The last three assessments are
Nutrition counseling. Nutritional state is an important baseline and outcome measures.
determinant of symptoms, disability, and prognosis in
COPD; both overweight and underweight can be a Several detailed questionnaires for assessing health
problem. Specific nutritional recommendations for status are available, including some that are specifically
patients with COPD are based on expert opinion and designed for patients with respiratory disease (e.g.,
some small randomized clinical trials209. Approximately Chronic Respiratory Disease Questionnaire57, St. George
25% of patients with Stage II: Moderate COPD to Stage Respiratory Questionnaire146), and there is increasing
IV: Very Severe COPD show a reduction in both their evidence that these questionnaires may be useful in a
body mass index and fat free mass135-137. A reduction in clinical setting. Health status can also be assessed by
body mass index is an independent risk factor for generic questionnaires, such as the Medical Outcomes
mortality in COPD patients138-140 (Evidence A). Study Short Form (SF36)147, to enable comparison of
quality of life in different diseases.
Health care workers should identify and correct the
reasons for reduced calorie intake in COPD patients. Economic cost of rehabilitation programs. A
Patients who become breathless while eating should be Canadian study showing statistically significant improve-
advised to take small, frequent meals. Poor dentition ments in dyspnea, fatigue, emotional health, and mastery
should be corrected and comorbidities (pulmonary sepsis, found that the incremental cost of pulmonary rehabilita-
lung tumors, etc.) should be managed appropriately. tion was $11,597 (CDN) per person148. A study from the
Improving the nutritional state of weight-losing COPD UK provided evidence that an intensive (6-week, 18-visit)
patients can lead to improved respiratory muscle multidisciplinary rehabilitation program was effective in
strength141-143. However, controversy remains as to decreasing use of health services122 (Evidence B).
whether this additional effort is cost effective141,142. Present Although there was no difference in the number of
evidence suggests that nutritional supplementa-tion alone hospital admissions between patients with disabling
may not be a sufficient strategy. Increased calorie intake COPD in a control group and those who participated in
is best accompanied by exercise regimes that have a the rehabilitation program, the number of days the
nonspecific anabolic action. This approach has not been rehabilitation group spent in the hospital was significantly
formally tested in large numbers of subjects. Anabolic lower. The rehabilitation group had more primary-care
steroids in patients with COPD with weight loss increase consultations at the general practitioner’s premises than
body weight and lean body mass but have little or no did the control group, but fewer primary-care home visits.
effect on exercise capacity144,145 Compared with the control group, the rehabilitation group
also showed greater improvements in walking ability and
Education. Most pulmonary rehabilitation programs in general and disease-specific health status.
include an educational component, but the specific
contributions of education to the improvements seen after Oxygen Therapy
pulmonary rehabilitation remain unclear.
Oxygen therapy, one of the principal non-pharmacologic
Assessment and follow-up. Baseline and outcome treatments for patients with Stage IV: Very Severe
assessments of each participant in a pulmonary COPD88,149, can be administered in three ways: long-term
rehabilitation program should be made to quantify continuous therapy, during exercise, and to relieve acute
individual gains and target areas for improvement. dyspnea. The primary goal of oxygen therapy is to
Assessments should include: increase the baseline PaO2 to at least 8.0 kPa (60 mm
Hg) at sea level and rest, and/or produce an SaO2 at
• Detailed history and physical examination. least 90%, which will preserve vital organ function by
• Measurement of spirometry before and after a ensuring adequate delivery of oxygen.
bronchodilator drug.
• Assessment of exercise capacity. The long-term administration of oxygen (> 15 hours per
• Measurement of health status and impact of day) to patients with chronic respiratory failure has
breathlessness. been shown to increase survival150,152. It can also have a
• Assessment of inspiratory and expiratory muscle beneficial impact on hemodynamics, hematologic
strength and lower limb strength (e.g., quadriceps) in characteristics, exercise capacity, lung mechanics, and
patients who suffer from muscle wasting. mental state151. Continuous oxygen therapy decreased
resting pulmonary artery pressure in one study150 but not

76 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 77

in another study152. Several controlled prospective long-term oxygen therapy, patients should be instructed
studies have shown that the primary hemodynamic effect to increase the flow by 1-2 L/min during the flight159.
of oxygen therapy is preventing the progression of Ideally, patients who fly should be able to maintain an
pulmonary hypertension153,154. Long-term oxygen therapy inflight PaO2 of at least 6.7 kPa (50 mm Hg). Studies
improves general alertness, motor speed, and hand grip, indicate that this can be achieved in those with moderate
although the data are less clear about changes in quality to severe hypoxemia at sea level by supplementary
of life and emotional state. The possibility of walking oxygen at 3 L/min by nasal cannulae or 31% by Venturi
while using some oxygen devices may help to improve facemask160. Those with a resting PaO2 at sea level of
physical conditioning and have a beneficial influence on > 9.3 kPa (70 mm Hg) are likely to be safe to fly without
the psychological state of patients155. supplementary oxygen159,161, although it is important to
emphasize that a resting PaO2 > 9.3 kPa (70 mm Hg) at
Long-term oxygen therapy is generally introduced in sea level does not exclude the development of severe
Stage IV: Very Severe COPD for patients who have: hypoxemia when travelling by air (Evidence C). Careful
consideration should be given to any comorbidity that
• PaO2 at or below 7.3 kPa (55 mm Hg) or SaO2 at or may impair oxygen delivery to tissues (e.g., cardiac
below 88%, with or without hypercapnia (Evidence A); or impairment, anemia). Also, walking along the aisle may
profoundly aggravate hypoxemia162.
• PaO2 between 7.3 kPa (55 mm Hg) and 8.0 kPa
(60 mm Hg), or SaO2 of 89%, if there is evidence of Ventilatory Support
pulmonary hypertension, peripheral edema
suggesting congestive cardiac failure, or polycythemia Although both noninvasive ventilation (using either
(hematocrit > 55%) (Evidence D). negative or positive pressure devices) and invasive
(conventional) mechanical ventilation are essentially
A decision about the use of long-term oxygen should be designed to manage and treat acute episodes of COPD,
based on the waking PaO2 values. The prescription for years noninvasive ventilation has been applied in
should always include the source of supplemental oxygen patients with Stage IV: Very Severe COPD and chronic
(gas or liquid), method of delivery, duration of use, and respiratory failure. This has followed the successful use of
flow rate at rest, during exercise, and during sleep. noninvasive ventilation in other forms of chronic
Oxygen therapy given during exercise increases walking respiratory failure due to chest wall deformities and/or
distance and endurance, optimizing oxygen delivery to neuromuscular disorders. Several scientific studies have
tissues and utilization by muscles. However, there are no examined the use of ventilatory support and there is no
data to suggest that long-term oxygen therapy changes convincing evidence that this therapy has a role in the
exercise capacity per se. Where available, this treatment management of stable COPD. It is possible that some
is usually restricted to patients who meet the criteria for patients with chronic hypercapnia may benefit from this
continuous oxygen therapy, or experience significant form of treatment, but no randomized controlled study
oxygen desaturation during exercise (Evidence C). has yet been reported.

Oxygen therapy reduces the oxygen cost of breathing Noninvasive mechanical ventilation. This modality of
and minute ventilation, a mechanism that although still ventilatory support is applied when endotracheal and
disputed helps to minimize the sensation of dyspnea. nasotracheal ventilation are not needed, using either
This has led to the use of short burst therapy to control negative pressure ventilation (nPV) or noninvasive
severe dyspnea such as occurs after climbing stairs. intermittent positive pressure ventilation (NIPPV).
However, there is no benefit from using short burst
oxygen for symptomatic relief before or after exercise210,211 Noninvasive negative pressure ventilation (nPV). The use
(Evidence B). of tank respirators, cuirass, or poncho ventilation is largely
of historical interest in COPD. Problems with patient
Cost considerations. Supplemental home oxygen is comfort and limited access restrict future use of nPV163,164.
usually the most costly component of outpatient therapy When this treatment is used in chronic respiratory failure,
for adults with COPD who require this therapy156. Studies some patients develop upper airway obstruction during
of the cost effectiveness of alternative outpatient oxygen sleep165. A comparison of domiciliary active and sham
delivery methods in the US and Europe suggest that nPV in patients with chronic respiratory failure due to
oxygen concentrator devices may be more cost effective COPD showed no differences in shortness of breath,
than cylinder delivery systems157,158. exercise tolerance, arterial blood gases, respiratory
muscle strength, or quality of life between the two
Oxygen use in air travel. Although air travel is safe for treatments166.
most patients with chronic respiratory failure who are on

MANAGEMENT OF COPD 77
GOLD_WR_05 8/18/05 12:56 PM Page 78

Noninvasive intermittent positive pressure ventilation Bullae may be removed to alleviate local symptoms
(NIPPV). The role of NIPPV in chronic respiratory failure such as hemoptysis, infection, or chest pain, and to allow
remains unsettled, although this is now the standard re-expansion of a compressed lung region. This is the
means of providing noninvasive ventilatory support in usual indication in patients with COPD. In considering the
other instances of chronic respiratory failure not directly possible benefit of surgery it is crucial to estimate the
related to COPD. NIPPV can be delivered by different effect of the bulla on the lung and the function of the
types of ventilators: volume-controlled, pressure-controlled, nonbullous lung. A thoracic CT scan, arterial blood gas
bilevel positive airway pressure, or continuous positive measurement, and comprehensive respiratory function
airway pressure. New devices with lower cost, greater tests are essential before making a decision regarding
ease of operation, and greater portability are constantly suitability for resection of a bulla. Normal or minimally
being developed167. Recent technical improvements have reduced diffusing capacity, absence of significant
facilitated the use of NIPPV while reducing the possibility hypoxemia, and evidence of regional reduction in perfusion
of air leaking through face or nasal masks. with good perfusion in the remaining lung are indications
a patient will likely benefit from surgery174. However,
A study of NIPPV compared to conventional therapy in a pulmonary hypertension, hypercapnia, and severe
population with end-stage COPD using a randomized, emphysema are not absolute contraindications for
crossover design for a 3-month period found that the bullectomy. Some investigators have recommended that
noninvasive approach is not well tolerated and is the bulla must occupy 50% or more of the hemithorax
associated with marginal clinical and functional and produce definite displacement of the adjacent lung
improvements168 (Evidence B). Although preliminary before surgery is performed175.
studies have suggested that combining NIPPV with
long-term oxygen therapy could be beneficial on certain Lung volume reduction surgery (LVRS). LVRS is a
outcome variables, based on a 12-month study169 and a surgical procedure in which parts of the lung are resected
24-month study170 in stable COPD patients with chronic to reduce hyperinflation176, making respiratory muscles
respiratory failure, its widespread use cannot be more effective pressure generators by improving their
advocated as yet171. However, compared with long-term mechanical efficiency (as measured by length/tension
oxygen therapy alone, the addition of NIPPV has some relationship, curvature of the diaphragm, and area of
effect on carbon dioxide retention and improved apposition)177,178. In addition, LVRS increases the elastic
shortness of breath170. recoil pressure of the lung and thus improves expiratory
flow rates179.
Given this conflicting evidence, long-term NIPPV cannot
be recommended for the routine treatment of patients LVRS does not improve life expectancy but improves
with chronic respiratory failure due to COPD. Nonethe- exercise capacity in patients with predominant upper lobe
less, the combination of NIPPV with long-term oxygen emphysema and a low post-rehabilitation exercise
therapy may be of some use in a selected subset of capacity212, and may improve global health status in
patients, particularly in those with pronounced daytime patients with heterogeneous emphysema213.
hypercapnia172.
In some centers with adequate experience, perioperative
Invasive (conventional) mechanical ventilation. mortality of LVRS has been reported to be less than 5%.
The appropriateness of using invasive (conventional) Results have been reported following bilateral (upper
ventilation in end-stage COPD continues to be debated. parts) resection using median sternotomy180,181 or
There are no guidelines to define which patients will video-assisted thoracoscopy (VATS)182. Most studies
benefit. select patients with FEV1 < 35% predicted, PaCO2
< 6.0 kPa (45 mm Hg), predominant upper lobe
Surgical Treatments emphysema on CT scan, and a residual volume of >
200% predicted. The average increase in FEV1 following
Bullectomy. Bullectomy is an older surgical procedure LVRS has ranged from 32% to 93%, and the decrease in
for bullous emphysema. By removing a large bulla that TLC from 15% to 20%180,183. LVRS appears to improve
does not contribute to gas exchange, the adjacent lung exercise capacity as well as quality of life in some patients.
parenchyma is decompressed. Bullectomy can be There are reports of these effects lasting more than one
performed thoracoscopically. In carefully selected year180-182. Patients with an FEV1 < 20 % predicted
patients, this procedure is effective in reducing dyspnea and either homogeneous emphysema on HRCT or a
and improving lung function173 (Evidence C). DLCO < 20 % predicted are at high risk for death after
LVRS and unlikely to benefit from the intervention184.

78 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 79

Hospital costs associated with LVRS in 52 consecutive poor general health status, age, obesity and COPD severity.
patients185 ranged from $11,712 to $121,829 (US). A comprehensive definition of postoperative pulmonary
Hospital charges in 23 consecutive patients admitted for complications should include only major pulmonary
LVRS at a single institution186 ranged from $20,032 to respiratory complications, namely lung infections,
$75,561 with a median charge of $26,669 (US). A small atelectasis and/or increased airflow obstruction, all
number of individuals incurred extraordinary costs potentially resulting in acute respiratory failure and
because of complications. Advanced age was a significant aggravation of underlying COPD214-219.
factor leading to higher expected total hospital costs.
Although there are some encouraging reports187, LVRS The incidence of increased risk of postoperative pulmonary
is still an experimental palliative surgical procedure188. complications in COPD patients may vary according to
Most results (Evidence C) reported to date are from the definition of postoperative pulmonary complications
uncontrolled studies; several large randomized multicenter and the severity of COPD, with relative ranges of the
studies are underway to investigate the effectiveness and order of 2.7 and 4.7214. The surgical site is the most
cost of LVRS in comparison to vigorous conventional important predictor, and risk increases as the incision
therapy189. Until the results of these controlled studies are approaches the diaphragm. Upper abdominal and thoracic
known, LVRS cannot be recommended for widespread surgery represents the greatest risk, the latter being
use. uncommon after interventions outside the thorax or
abdomen. Most reports conclude that epidural or spinal
Lung transplantation. In appropriately selected patients anesthesia have a lower risk than with general anesthesia,
with very advanced COPD, lung transplantation has although the results are not totally uniform.
been shown to improve quality of life and functional
capacity190-193 (Evidence C), although the Joint United Patient-risk factors are identified by careful history, physical
examination, chest radiography, and pulmonary function
Network for Organ Sharing in 1998 found that lung
tests. Although the value of pulmonary function tests
transplantation does not confer a survival benefit in
remains contentious, there is consensus that all COPD
patients with end-stage emphysema after two years192.
candidates for lung resection should undergo a complete
Criteria for referral for lung transplantation include FEV1
battery, including forced spirometry with bronchodilator
< 35% predicted, PaO2 < 7.3-8.0 kPa (55-60 mm Hg), response, static lung volumes, diffusing capacity and
PaCO2 > 6.7 kPa (50 mm Hg), and secondary pulmonary arterial blood gases at rest. One theoretical rationale
hypertension194,195. behind the assessment of pulmonary function measurement
is the identification of COPD patients in whom the risk is
Lung transplantation is limited by the shortage of donor so elevated that surgery should be contraindicated.
organs, which has led some centers to adopt the single
lung technique. The common complications seen in Several studies in high risk COPD patients suggest that
COPD patients after lung transplantation, apart from there is threshold beyond which the risk of surgery is
operative mortality, are acute rejection and bronchiolitis prohibitive. The risk of postoperative respiratory failure
obliterans, CMV, other opportunistic fungal (Candida, appears to be in patients undergoing pneumonectomy
Aspergillus, Cryptococcus, Carini) or bacterial with a preoperative FEV1 < 2 L or 50% predicted and/or
(Pseudomonas, Staphylococcus species) infections, a DLCO < 50% predicted218. COPD patients at high risk
lymphoproliferative disease, and lymphomas191. due to poor lung function should undergo further lung
function assessment, for example, regional distribution
Another limitation of lung transplantation is its cost. of perfusion and exercise capacity219. To prevent
Hospitalization costs associated with lung transplantation postoperative pulmonary complications, stable COPD
have ranged from $110,000 to well over $200,000 patients clinically symptomatic and/or with limited exercise
(US). Costs remain elevated for months to years after capacity should be treated, before surgery, intensely with
surgery due to the high cost of complications and the all the measures already well established for stable
immunosuppressive regimens196-199 that must be initiated COPD patients who are not about to have surgery.
during or immediately after surgery. Surgery should be postponed if an exacerbation is present.

Special Considerations Surgery in patients with COPD needs to be differentiated


from that aimed to improve function and symptoms for
Surgery in COPD. Postoperative pulmonary complications COPD. This includes bullectomy, lung volume reduction
are as important and common as postoperative cardiac surgery and lung transplantation219.
complications and, consequently, are a key component
of increased risk of surgery in COPD patients. The principal
potential factors contributing to the risk include smoking,

MANAGEMENT OF COPD 79
GOLD_WR_05 8/18/05 12:56 PM Page 80

REFERENCES 16. Berger R, Smith D. Effect of inhaled metaproterenol on


exercise performance in patients with stable "fixed" airway
1. Reis AL. Response to bronchodilators. In: Clausen J, ed. obstruction. Am Rev Respir Dis 1988; 138:624-9.
Pulmonary function testing: guidelines and controversies. 17. Hay JG, Stone P, Carter J, Church S, Eyre-Brook A,
New York: Academic Press, 1982. Pearson MG, et al. Bronchodilator reversibility, exercise
2. Janelli LM, Scherer YK, Schmieder LE. Can a pulmonary performance and breathlessness in stable chronic obstruc-
health teaching program alter patients' ability to cope with tive pulmonary disease. Eur Respir J 1992; 5:659-64.
COPD? Rehabil Nurs 1991; 16:199-202. 18. Vathenen AS, Britton JR, Ebden P, Cookson JB, Wharrad
3. Ashikaga T, Vacek PM, Lewis SO. Evaluation of a commu- HJ, Tattersfield AE. High-dose inhaled albuterol in severe
nity-based education program for individuals with chronic chronic airflow limitation. Am Rev Respir Dis 1988; 138:850-5.
obstructive pulmonary disease. J Rehabil 1980; 46:23-7. 19. Gross NJ, Petty TL, Friedman M, Skorodin MS, Silvers
4. Toshima MT, Kaplan RM, Ries AL. Experimental evaluation GW, Donohue JF. Dose response to ipratropium as a nebu-
of rehabilitation in chronic obstructive pulmonary disease: lized solution in patients with chronic obstructive pul-
short-term effects on exercise endurance and health sta- monary disease. A three-center study. Am Rev Respir Dis
tus. Health Psychol 1990; 9:237-52. 1989; 139:1188-91.
5. Celli BR. Pulmonary rehabilitation in patients with COPD. 20. Chrystyn H, Mulley BA, Peake MD. Dose response relation
Am J Respir Crit Care Med 1995; 152:861-4. to oral theophylline in severe chronic obstructive airways
6. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey disease. BMJ 1988; 297:1506-10.
WC, Buist AS, et al. Effects of smoking intervention and 21. Higgins BG, Powell RM, Cooper S, Tattersfield AE. Effect
the use of an inhaled anticholinergic bronchodilator on the of salbutamol and ipratropium bromide on airway calibre
rate of decline of FEV1. The Lung Health Study. JAMA and bronchial reactivity in asthma and chronic bronchitis.
1994; 272:1497-505. Eur Respir J 1991; 4:415-20.
7. Stewart MA. Effective physician-patient communication and 22. Ericsson CH, Svartengren K, Svartengren M, Mossberg B,
health outcomes: a review. CMAJ 1995; 152:1423-33. Philipson K, Blomquist M, et al. Repeatability of airway
8. Clark NM, Nothwehr F, Gong M, Evans D, Maiman LA, deposition and tracheobronchial clearance rate over three
Hurwitz ME, et al. Physician-patient partnership in manag- days in chronic bronchitis. Eur Respir J 1995; 8:1886-93.
ing chronic illness. Acad Med 1995; 70:957-9. 23. Kim CS, Kang TC. Comparative measurement of lung
9. Heffner JE, Fahy B, Hilling L, Barbieri C. Outcomes of deposition of inhaled fine particles in normal subjects and
advance directive education of pulmonary rehabilitation patients with obstructive airway disease. Am J Respir Crit
patients. Am J Respir Crit Care Med 1997; 155:1055-9. Care Med 1997; 155:899-905.
10. Tougaard L, Krone T, Sorknaes A, Ellegaard H. Economic 24. Boe J, Dennis JH, O'Driscoll BR, Bauer TT, Carone M,
benefits of teaching patients with chronic obstructive pul- Dautzenberg B, et al. European Respiratory Society
monary disease about their illness. The PASTMA Group. Guidelines on the use of nebulizers. Eur Respir J 2001;
Lancet 1992; 339:1517-20. 18(1):228-42.
11. Pauwels RA, Lofdahl CG, Laitinen LA, Schouten JP, 25. O'Driscoll BR, Kay EA, Taylor RJ, Weatherby H, Chetty
Postma DS, Pride NB, et al. Long-term treatment with MC, Bernstein A. A long-term prospective assessment of
inhaled budesonide in persons with mild chronic obstruc- home nebulizer treatment. Respir Med 1992; 86:317-25.
tive pulmonary disease who continue smoking. European 26. Jenkins SC, Heaton RW, Fulton TJ, Moxham J.
Respiratory Society Study on Chronic Obstructive Comparison of domiciliary nebulized salbutamol and salbu-
Pulmonary Disease. N Engl J Med 1999; 340:1948-53. tamol from a metered-dose inhaler in stable chronic airflow
12. Vestbo J, Sorensen T, Lange P, Brix A, Torre P, Viskum K. limitation. Chest 1987; 91:804-7.
Long-term effect of inhaled budesonide in mild and moder- 27. COMBIVENT Inhalation Aerosol Study Group. In chronic
ate chronic obstructive pulmonary disease: a randomised obstructive pulmonary disease, a combination of ipratropi-
controlled trial. Lancet 1999; 353:1819-23. um and albuterol is more effective than either agent alone.
13. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson An 85-day multicenter trial. Chest 1994; 105:1411-9.
JA, Maslen TK. Randomised, double blind, placebo con- 28. van Schayck CP, Folgering H, Harbers H, Maas KL, van
trolled study of fluticasone propionate in patients with mod- Weel C. Effects of allergy and age on responses to salbu-
erate to severe chronic obstructive pulmonary disease: the tamol and ipratropium bromide in moderate asthma and
ISOLDE trial. BMJ 2000; 320:1297-303. chronic bronchitis. Thorax 1991; 46:355-9.
14. Calverley PMA. Symptomatic bronchodilator treatment. In: 29. Ulrik CS. Efficacy of inhaled salmeterol in the management
Calverley PMA, Pride NB, eds. Chronic obstructive pul- of smokers with chronic obstructive pulmonary disease: a
monary disease. London: Chapman and Hall, 1995:419-445. single centre randomised, double blind, placebo controlled,
15. Belman MJ, Botnick WC, Shin JW. Inhaled bronchodilators crossover study. Thorax 1995; 50:750-4.
reduce dynamic hyperinflation during exercise in patients 30. Boyd G, Morice AH, Pounsford JC, Siebert M, Peslis N,
with chronic obstructive pulmonary disease. Am J Respir Crawford C. An evaluation of salmeterol in the treatment of
Crit Care Med 1996; 153:967-75. chronic obstructive pulmonary disease (COPD). Eur Respir
J 1997; 10:815-21.

80 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 81

31. Cazzola M, Matera MG, Santangelo G, Vinciguerra A, 45. Disse B, Speck GA, Rominger KL, Witek TJ, Jr., Hammer
Rossi F, D'Amato G. Salmeterol and formoterol in partially R. Tiotropium (Spiriva): mechanistical considerations and
reversible severe chronic obstructive pulmonary disease: a clinical profile in obstructive lung disease. Life Sci 1999;
dose-response study. Respir Med 1995; 89:357-62. 64:457-64.
32. Rossi A, Kristufek P, Levine BE, Thomson MH, Till D, 46. Anthonisen NR, Connett JE, Enright PL, Manfreda J.
Kottakis J, et al. Comparison of the efficacy, tolerability, Hospitalizations and mortality in the Lung Health Study.
and safety of formoterol dry powder and oral, slow-release Am J Respir Crit Care Med 2002; 166:333-9.
theophylline in the treatment of COPD. Chest 2002; 47. Aubier M. Pharmacotherapy of respiratory muscles. Clin
121:1058-69. Chest Med 1988; 9:311-24.
33. van Noord JA, Bantje TA, Eland ME, Korducki L, 48. Moxham J. Aminophylline and the respiratory muscles: an
Cornelissen PJ. A randomised controlled comparison of alternative view. Clin Chest Med 1988; 9:325-36.
tiotropium nd ipratropium in the treatment of chronic 49. Murciano D, Auclair MH, Pariente R, Aubier M. A random-
obstructive pulmonary disease. The Dutch Tiotropium ized, controlled trial of theophylline in patients with severe
Study Group. Thorax 2000; 55:289-94. chronic obstructive pulmonary disease. N Engl J Med
34. Vincken W, van Noord JA, Greefhorst AP, Bantje TA, 1989; 320:1521-5.
Kesten S, Korducki L, et al. Improved health outcomes in 50. McKay SE, Howie CA, Thomson AH, Whiting B, Addis GJ.
patients with COPD during 1 yr's treatment with tiotropium. Value of theophylline treatment in patients handicapped by
Eur Respir J 2002; 19:209-16. chronic obstructive lung disease. Thorax 1993; 48:227-32.
35. Casaburi R, Mahler DA, Jones PW, Wanner A, San PG, 51. Taylor DR, Buick B, Kinney C, Lowry RC, McDevitt DG.
ZuWallack RL, et al. A long-term evaluation of once-daily The efficacy of orally administered theophylline, inhaled
inhaled tiotropium in chronic obstructive pulmonary dis- salbutamol, and a combination of the two as chronic thera-
ease. Eur Respir J 2002; 19:217-24. py in the management of chronic bronchitis with reversible
36. Ikeda A, Nishimura K, Koyama H, Izumi T. Bronchodilating air-flow obstruction. Am Rev Respir Dis 1985; 131:747-51.
effects of combined therapy with clinical dosages of iprat- 52. The COMBIVENT Inhalation Solution Study Group.
ropium bromide and salbutamol for stable COPD: compari- Routine nebulized ipratropium and albuterol together are
son with ipratropium bromide alone. Chest 1995; 107:401-5. better than either alone in COPD. Chest 1997; 112:1514-21.
37. Guyatt GH, Townsend M, Pugsley SO, Keller JL, Short HD, 53. Gross N, Tashkin D, Miller R, Oren J, Coleman W, Linberg
Taylor DW, et al. Bronchodilators in chronic air-flow limita- S. Inhalation by nebulization of albuterol-ipratropium com-
tion. Effects on airway function, exercise capacity, and bination (Dey combination) is superior to either agent
quality of life. Am Rev Respir Dis 1987; 135:1069-74. alone in the treatment of chronic obstructive pulmonary
38. Mahler DA, Donohue JF, Barbee RA, Goldman MD, Gross disease. Dey Combination Solution Study Group.
NJ, Wisniewski ME, et al. Efficacy of salmeterol xinafoate Respiration 1998; 65:354-62.
in the treatment of COPD. Chest 1999; 115:957-65. 54. van Noord JA, de Munck DR, Bantje TA, Hop WC, Akveld
39. Dahl R, Greefhorst LA, Nowak D, Nonikov V, Byrne AM, ML, Bommer AM. Long-term treatment of chronic obstruc-
Thomson MH, et al. Inhaled formoterol dry powder versus tive pulmonary disease with salmeterol and the additive
ipratropium bromide in chronic obstructive pulmonary dis- effect of ipratropium. Eur Respir J 2000; 15:878-85.
ease. Am J Respir Crit Care Med 2001; 164:778-84. 55. ZuWallack RL, Mahler DA, Reilly D, Church N, Emmett A,
40. Shim CS, Williams MH, Jr. Bronchodilator response to oral Rickard K, et al. Salmeterol plus theophylline combination
aminophylline and terbutaline versus aerosol albuterol in therapy in the treatment of COPD. Chest 2001; 119:1661-70.
patients with chronic obstructive pulmonary disease. Am J 56. Bellia V, Foresi A, Bianco S, Grassi V, Olivieri D, Bensi G,
Med 1983; 75:697-701. et al. Efficacy and safety of oxitropium bromide, theo-
41. Lipworth BJ, McDevitt DG, Struthers AD. Hypokalemic and phylline and their combination in COPD patients: a double-
ECG sequelae of combined beta-agonist/diuretic therapy. blind, randomized, multicentre study (BREATH Trial).
Protection by conventional doses of spironolactone but not Respir Med 2002; 96:881-9.
triamterene. Chest 1990; 98:811-5. 57. Guyatt GH, Berman LB, Townsend M, Pugsley SO,
42. Uren NG, Davies SW, Jordan SL, Lipkin DP. Inhaled bron- Chambers LW. A measure of quality of life for clinical trials
chodilators increase maximum oxygen consumption in in chronic lung disease. Thorax 1987; 42:773-8.
chronic left ventricular failure. Eur Heart J 1993; 14:744-50. 58. Callahan CM, Dittus RS, Katz BP. Oral corticosteroid thera-
43. Khoukaz G, Gross NJ. Effects of salmeterol on arterial py for patients with stable chronic obstructive pulmonary
blood gases in patients with stable chronic obstructive pul- disease. A meta-analysis. Ann Intern Med 1991; 114:216-23.
monary disease. Comparison with albuterol and ipratropi- 59. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson
um. Am J Respir Crit Care Med 1999; 160:1028-30. JA. Prednisolone response in patients with chronic
44. Barnes PJ. Bronchodilators: basic pharmacology. In: obstructive pulmonary disease: results from the ISOLDE
Calverley PMA, Pride NB, eds. Chronic obstructive pul- study. Thorax. 2003 Aug;58(8):654-8.
monary disease. London: Chapman and Hall, 1995:391-417. 60. Postma DS, Peters I, Steenhuis EJ, Sluiter HJ. Moderately
severe chronic airflow obstruction. Can corticosteroids slow
down obstruction? Eur Respir J 1988; 1:22-6.

MANAGEMENT OF COPD 81
GOLD_WR_05 8/18/05 12:56 PM Page 82

61. Postma DS, Steenhuis EJ, van der Weele LT, Sluiter HJ. 75. Simberkoff MS, Cross AP, Al-Ibrahim M, Baltch AL,
Severe chronic airflow obstruction: can corticosteroids slow Geiseler PJ, Nadler J, et al. Efficacy of pneumococcal vac-
down progression? Eur J Respir Dis 1985; 67:56-64. cine in high-risk patients. Results of a Veterans Administration
62. Decramer M, de Bock V, Dom R. Functional and histologic Cooperative Study. N Engl J Med 1986; 315:1318-27.
picture of steroid-induced myopathy in chronic obstructive 76. Williams JH, Jr., Moser KM. Pneumococcal vaccine and
pulmonary disease. Am J Respir Crit Care Med 1996; patients with chronic lung disease. Ann Intern Med 1986;
153:1958-64. 104:106-9.
63. Decramer M, Lacquet LM, Fagard R, Rogiers P. Corticosteroids 77. Davis AL, Aranda CP, Schiffman G, Christianson LC.
contribute to muscle weakness in chronic airflow obstruc- Pneumococcal infection and immunologic response to
tion. Am J Respir Crit Care Med 1994; 150:11-6. pneumococcal vaccine in chronic obstructive pulmonary
64. Decramer M, Stas KJ. Corticosteroid-induced myopathy disease. A pilot study. Chest 1987; 92:204-12.
involving respiratory muscles in patients with chronic 78. Francis RS, May JR, Spicer CC. Chemotherapy of bronchi-
obstructive pulmonary disease or asthma. Am Rev Respir tis: influence of penicillin and tetracylcline administered daily,
Dis 1992; 146:800-2. or intermittently for exacerbations. BMJ 1961; 2:979-985.
65. The Lung Health Study Research Group. Effect of inhaled 79. Francis RS, Spicer CC. Chemotherapy in chronic bronchi-
triamcinolone on the decline in pulmonary function in tis: influence of daily penicillin and teracycline on exacer-
chronic obstructive pulmonary disease: Lung Health Study bations and their cost. A report to the research committee
II. N Engl J Med 2000; 343:1902-1909. of the British Tuberculosis Assoication by their Chronic
66. Mahler DA, Wire P, Horstman D, Chang CN, Yates J, Bronchitis subcommittee. BMJ 1960; 1:297-303.
Fischer T, et al. Effectiveness of fluticasone propionate and 80. Fletcher CM, Ball JD, Carstairs LW, Couch AHC, Crofton JM,
Edge JR, et al. Value of chemoprophylaxis and chemotherapy
salmeterol combination delivered via the Diskus device in
in early chronic bronchitis. A report to the Medical Research
the treatment of chronic obstructive pulmonary disease.
Council by their Working Party on trials of chemotherpay in
Am J Respir Crit Care Med 2002; 166(8):1084-91.
early chronic bronchitis. BMJ 1966; 1(5499):1317-1322.
67. Jones PW, Willits LR, Burge PS, Calverley PM. Disease
81. Johnston RN, McNeill RS, Smith DH, Dempster MB, Nairn
severity and the effect of fluticasone propionate on chronic
JR, Purvis MS, et al. Five-year winter chemoprophylaxis for
obstructive pulmonary disease exacerbations. Eur Respir J
chronic bronchitis. Br Med J 1969; 4:265-9.
2003; 21:68-73. 82. Isada CM, Stoller JK. Chronic bronchitis: the role of antibi-
68. Calverley P, Pauwels R, Vestbo J, Jones P, Pride N, otics. In: Niederman MS, Sarosi GA, Glassroth J, eds.
Gulsvik A, et al. Combined salmeterol and fluticasone in Respiratory infections: a scientific basis for management.
the treatment of chronic obstructive pulmonary disease: a London: WB Saunders, 1994:621-33.
randomised controlled trial. Lancet 2003; 361:449-56. 83. Siafakas NM, Bouros D. Management of acute exacerba-
69. Szafranski W, Cukier A, Ramirez A, Menga G, Sansores R, tion of chronic obstructive pulmonary disease. In: Postma
Nahabedian S, et al. Efficacy and safety of budesonide/for- DS, Siafakas NM, eds. Management of chronic obstructive
moterol in the management of chronic obstructive pul- pulmonary disease. Sheffield: ERS Monograph, 1998:264-77.
monary disease. Eur Respir J 2003; 21:74-81. 84. Allegra L, Cordaro CI, Grassi C. Prevention of acute exac-
70. Johnell O, Pauwels R, Lofdahl CG, Laitinen LA, Postma erbations of chronic obstructive bronchitis with carbocys-
DS, Pride NB, et al. Bone mineral density in patients with teine lysine salt monohydrate: a multicenter, double- blind,
chronic obstructive pulmonary disease treated with budes- placebo-controlled trial. Respiration 1996; 63:174-80.
onide Turbuhaler. Eur Respir J 2002; 19:1058-63. 85. Guyatt GH, Townsend M, Kazim F, Newhouse MT. A con-
71. NHLBI/WHO workshop report: global strategy for asthma trolled trial of ambroxol in chronic bronchitis. Chest 1987;
management and prevention. Washington, D.C.: 92:618-20.
Department of Health and Human Services, 2002:NIH 86. Petty TL. The National Mucolytic Study. Results of a ran-
Publication No. 02-2659. domized, double-blind, placebo-controlled study of iodinated
72. Nichol KL, Margolis KL, Wuorenma J, Von Sternberg T. The glycerol in chronic obstructive bronchitis. Chest 1990; 97:75-83.
efficacy and cost effectiveness of vaccination against 87. Poole PJ, Black PN. Mucolytic agents for chronic bronchitis
influenza among elderly persons living in the community. N or chronic obstructive pulmonary disease. Cochrane
Engl J Med 1994; 331:778-84. Database Syst Rev 2000; 2:Available from URL
73. Edwards KM, Dupont WD, Westrich MK, Plummer WD, Jr., www.update-software.com or www.updatusa.com.
Palmer PS, Wright PF. A randomized controlled trial of 88. Siafakas NM, Vermeire P, Pride NB, Paoletti P, Gibson J,
cold-adapted and inactivated vaccines for the prevention of Howard P, et al. Optimal assessment and management of
influenza A disease. J Infect Dis 1994; 169:68-76. chronic obstructive pulmonary disease (COPD). The
74. Hak E, van Essen GA, Buskens E, Stalman W, de Melker European Respiratory Society Task Force. Eur Respir J
RA. Is immunising all patients with chronic lung disease in 1995; 8:1398-420.
the community against influenza cost effective? Evidence 89. American Thoracic Society. Standards for the diagnosis
from a general practice based clinical prospective cohort and care of patients with chronic obstructive pulmonary
study in Utrecht, The Netherlands. J Epidemiol Community disease (COPD) and asthma. Adopted by the ATS Board
of Directors, November 1986. Am Rev Respir Dis 1987;
Health 1998; 52:120-5.
136:225-44.

82 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 83

90. Hansen NC, Skriver A, Brorsen-Riis L, Balslov S, Evald T, 105. Woodcock AA, Gross ER, Gellert A, Shah S, Johnson M,
Maltbaek N, et al. Orally administered N-acetylcysteine Geddes DM. Effects of dihydrocodeine, alcohol, and caf-
may improve general well-being in patients with mild feine on breathlessness and exercise tolerance in patients
chronic bronchitis. Respir Med 1994; 88:531-5. with chronic obstructive lung disease and normal blood
91. British Thoracic Society Research Committee. Oral N- gases. N Engl J Med 1981; 305:1611-6.
acetylcysteine and exacerbation rates in patients with 106. Rice KL, Kronenberg RS, Hedemark LL, Niewoehner DE.
chronic bronchitis and severe airways obstruction. Thorax Effects of chronic administration of codeine and promet-
1985; 40:832-5. hazine on breathlessness and exercise tolerance in
92. Boman G, Backer U, Larsson S, Melander B, Wahlander L. patients with chronic airflow obstruction. Br J Dis Chest
Oral acetylcysteine reduces exacerbation rate in chronic 1987; 81:287-92.
bronchitis: report of a trial organized by the Swedish 107. Poole PJ, Veale AG, Black PN. The effect of sustained-
Society for Pulmonary Diseases. Eur J Respir Dis 1983; release morphine on breathlessness and quality of life in
64:405-15. severe chronic obstructive pulmonary disease. Am J
93. Rasmussen JB, Glennow C. Reduction in days of illness Respir Crit Care Med 1998; 157:1877-80.
after long-term treatment with N- acetylcysteine controlled- 108. American Thoracic Society. Pulmonary rehabilitation-1999.
release tablets in patients with chronic bronchitis. Eur Am J Respir Crit Care Med 1999; 159:1666-82.
Respir J 1988; 1:351-5. 109. Fishman AP. Pulmonary rehabilitation research. Am J
94. Collet JP, Shapiro P, Ernst P, Renzi T, Ducruet T, Robinson Respir Crit Care Med 1994; 149:825-33.
A. Effects of an immunostimulating agent on acute exacer- 110. Pulmonary rehabilitation: joint ACCP/AACVPR evidence-
bations and hospitalizations in patients with chronic based guidelines. ACCP/AACVPR Pulmonary
obstructive pulmonary disease. The PARI-IS Study Rehabilitation Guidelines Panel. American College of
Steering Committee and Research Group. Prevention of Chest Physicians. American Association of Cardiovascular
Acute Respiratory Infection by an Immunostimulant. Am J and Pulmonary Rehabilitation. Chest 1997; 112:1363-96.
Respir Crit Care Med 1997; 156:1719-24. 111. Lacasse Y, Wong E, Guyatt GH, King D, Cook DJ, Goldstein
95. Anthonisen NR. OM-8BV for COPD. Am J Respir Crit Care RS. Meta-analysis of respiratory rehabilitation in chronic
Med 1997; 156:1713-4. obstructive pulmonary disease. Lancet 1996; 348:1115-9.
96. Irwin RS, Boulet LP, Cloutier MM, Fuller R, Gold PM, 112. Goldstein RS, Gort EH, Stubbing D, Avendano MA, Guyatt
Hoffstein V, et al. Managing cough as a defense mecha- GH. Randomised controlled trial of respiratory rehabilita-
nism and as a symptom. A consensus panel report of the tion. Lancet 1994; 344:1394-7.
American College of Chest Physicians. Chest 1998; 113. Wijkstra PJ, Van Altena R, Kraan J, Otten V, Postma DS,
114:133S-181S. Koeter GH. Quality of life in patients with chronic obstruc-
97. Barbera JA, Roger N, Roca J, Rovira I, Higenbottam TW, tive pulmonary disease improves after rehabilitation at
Rodriguez-Roisin R. Worsening of pulmonary gas home. Eur Respir J 1994; 7:269-73.
exchange with nitric oxide inhalation in chronic obstructive 114. O'Donnell DE, McGuire M, Samis L, Webb KA. The impact
pulmonary disease. Lancet 1996; 347:436-40. of exercise reconditioning on breathlessness in severe
98. Jones AT, Evans TW. NO: COPD and beyond. Thorax chronic airflow limitation. Am J Respir Crit Care Med 1995;
1997; 52 Suppl 3:S16-21. 152:2005-13.
99. Watanabe S, Kanner RE, Cutillo AG, Menlove RL, Bachand 115. Lake FR, Henderson K, Briffa T, Openshaw J, Musk AW.
RT, Jr., Szalkowski MB, et al. Long-term effect of almitrine Upper-limb and lower-limb exercise training in patients with
bismesylate in patients with hypoxemic chronic obstructive chronic airflow obstruction. Chest 1990; 97:1077-82.
pulmonary disease. Am Rev Respir Dis 1989; 140:1269-73. 116. Ries AL, Ellis B, Hawkins RW. Upper extremity exercise
100. Bardsley PA, Howard P, DeBacker W, Vermeire P, Mairesse training in chronic obstructive pulmonary disease. Chest
M, Ledent C, et al. Two years treatment with almitrine 1988; 93:688-92.
bismesylate in patients with hypoxic chronic obstructive air- 117. Martinez FJ, Vogel PD, Dupont DN, Stanopoulos I, Gray A,
ways disease. Eur Respir J 1991; 4:308-10. Beamis JF. Supported arm exercise vs unsupported arm
101. Winkelmann BR, Kullmer TH, Kneissl DG, Trenk D, exercise in the rehabilitation of patients with severe chronic
Kronenberger H. Low-dose almitrine bismesylate in the airflow obstruction. Chest 1993; 103:1397-402.
treatment of hypoxemia due to chronic obstructive pul- 118. Wijkstra PJ, Ten Vergert EM, van Altena R, Otten V, Kraan
monary disease. Chest 1994; 105:1383-91. J, Postma DS, et al. Long term benefits of rehabilitation at
102. Jennings AL, Davies AN, Higgins JP, Gibbs JS, Broadley home on quality of life and exercise tolerance in patients
KE. A systematic review of the use of opioids in the man- with chronic obstructive pulmonary disease. Thorax 1995;
agement of dyspnoea. Thorax 2002; 57:939-44. 50:824-8.
103. Eiser N, Denman WT, West C, Luce P. Oral diamorphine: 119. Berry MJ, Rejeski WJ, Adair NE, Zaccaro D. Exercise reha-
lack of effect on dyspnoea and exercise tolerance in the bilitation and chronic obstructive pulmonary disease stage.
"pink puffer" syndrome. Eur Respir J 1991; 4:926-31. Am J Respir Crit Care Med 1999; 160:1248-53.
104. Young IH, Daviskas E, Keena VA. Effect of low dose nebu-
lised morphine on exercise endurance in patients with
chronic lung disease. Thorax 1989; 44:387-90.

MANAGEMENT OF COPD 83
GOLD_WR_05 8/18/05 12:56 PM Page 84

120. Foglio K, Bianchi L, Bruletti G, Battista L, Pagani M, 135. Schols AM, Soeters PB, Dingemans AM, Mostert R,
Ambrosino N. Long-term effectiveness of pulmonary reha- Frantzen PJ, Wouters EF. Prevalence and characteristics of
bilitation in patients with chronic airway obstruction. Eur nutritional depletion in patients with stable COPD eligible
Respir J 1999; 13:125-32. for pulmonary rehabilitation. Am Rev Respir Dis 1993;
121. Young P, Dewse M, Fergusson W, Kolbe J. Improvements 147:1151-6.
in outcomes for chronic obstructive pulmonary disease 136. Engelen MP, Schols AM, Baken WC, Wesseling GJ,
(COPD) attributable to a hospital-based respiratory rehabil- Wouters EF. Nutritional depletion in relation to respiratory
itation programme. Aust N Z J Med 1999; 29:59-65. and peripheral skeletal muscle function in out-patients with
122. Griffiths TL, Burr ML, Campbell IA, Lewis-Jenkins V, COPD. Eur Respir J 1994; 7:1793-7.
Mullins J, Shiels K, et al. Results at 1 year of outpatient 137. Wilson DO, Rogers RM, Wright EC, Anthonisen NR. Body
multidisciplinary pulmonary rehabilitation: a randomised weight in chronic obstructive pulmonary disease. The
controlled trial. Lancet 2000; 355:362-8. National Institutes of Health Intermittent Positive-Pressure
123. McGavin CR, Gupta SP, Lloyd EL, McHardy GJ. Physical Breathing Trial. Am Rev Respir Dis 1989; 139:1435-8.
rehabilitation for the chronic bronchitic: results of a con- 138. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss
trolled trial of exercises in the home. Thorax 1977; 32:307-11. is a reversible factor in the prognosis of chronic obstructive
124. Wedzicha JA, Bestall JC, Garrod R, Garnham R, Paul EA, pulmonary disease. Am J Respir Crit Care Med 1998;
Jones PW. Randomized controlled trial of pulmonary reha- 157:1791-7.
bilitation in severe chronic obstructive pulmonary disease 139. Gray-Donald K, Gibbons L, Shapiro SH, Macklem PT,
patients, stratified with the MRC dyspnoea scale. Eur Martin JG. Nutritional status and mortality in chronic
Respir J 1998; 12:363-9. obstructive pulmonary disease. Am J Respir Crit Care Med
125. Singh SJ, Morgan MD, Scott S, Walters D, Hardman AE. 1996; 153:961-6.
Development of a shuttle walking test of disability in 140. Gorecka D, Gorzelak K, Sliwinski P, Tobiasz M, Zielinski J.
patients with chronic airways obstruction. Thorax 1992; Effect of long-term oxygen therapy on survival in patients
47:1019-24. with chronic obstructive pulmonary disease with moderate
126. Mahler DA. Pulmonary rehabilitation. Chest 1998; hypoxaemia. Thorax 1997; 52:674-9.
113:263S-268S. 141. Efthimiou J, Fleming J, Gomes C, Spiro SG. The effect of
127. American College of Sports Medicine position stand. The supplementary oral nutrition in poorly nourished patients
recommended quantity and quality of exercise for develop- with chronic obstructive pulmonary disease. Am Rev
ing and maintaining cardiorespiratory and muscular fitness Respir Dis 1988; 137:1075-82.
in healthy adults. Med Sci Sports Exerc 1990; 22:265-74. 142. Rogers RM, Donahoe M, Costantino J. Physiologic effects
128. Behnke M, Taube C, Kirsten D, Lehnigk B, Jorres RA, of oral supplemental feeding in malnourished patients with
Magnussen H. Home-based exercise is capable of pre- chronic obstructive pulmonary disease. A randomized con-
serving hospital-based improvements in severe chronic trol study. Am Rev Respir Dis 1992; 146:1511-7.
obstructive pulmonary disease. Respir Med 2000; 94:1184-91. 143. Whittaker JS, Ryan CF, Buckley PA, Road JD. The effects
129. Finnerty JP, Keeping I, Bullough I, Jones J. The effective- of refeeding on peripheral and respiratory muscle function
ness of outpatient pulmonary rehabilitation in chronic lung in malnourished chronic obstructive pulmonary disease
disease: a randomized controlled trial. Chest 2001; patients. Am Rev Respir Dis 1990; 142:283-8.
119:1705-10. 144. Yeh SS, DeGuzman B, Kramer T. Reversal of COPD-asso-
130. Green RH, Singh SJ, Williams J, Morgan MD. A ran- ciated weight loss using the anabolic agent oxandrolone.
domised controlled trial of four weeks versus seven weeks Chest 2002; 122:421-8.
of pulmonary rehabilitation in chronic obstructive pul- 145. Weisberg J, Wanger J, Olson J, Streit B, Fogarty C, Martin
monary disease. Thorax 2001; 56:143-5. T, et al. Megestrol acetate stimulates weight gain and ven-
131. Ries AL, Kaplan RM, Myers R, Prewitt LM. Maintenance tilation in underweight COPD patients. Chest 2002;
after pulmonary rehabilitation in chronic lung disease: a 121:1070-8.
randomized trial. Am J Respir Crit Care Med 2003; 146. Jones PW, Quirk FH, Baveystock CM. The St George's
167:880-8. Respiratory Questionnaire. Respir Med 1991; 85 Suppl
132. Belman MJ, Botnick WC, Nathan SD, Chon KH. Ventilatory B:25-31; discussion 33-7.
load characteristics during ventilatory muscle training. Am 147. Ware JE, Jr., Sherbourne CD. The MOS 36-item short-form
J Respir Crit Care Med 1994; 149:925-9. health survey (SF-36). I. Conceptual framework and item
133. Lotters F, van Tol B, Kwakkel G, Gosselink R. Effects of selection. Med Care 1992; 30:473-83.
controlled inspiratory muscle training in patients with 148. Goldstein RS, Gort EH, Guyatt GH, Feeny D. Economic
COPD: a meta-analysis. Eur Respir J 2002; 20:570-6. analysis of respiratory rehabilitation. Chest 1997; 112:370-9.
134. Bernard S, Whittom F, Leblanc P, Jobin J, Belleau R, 149. American Thoracic Society. Standards for the diagnosis
Berube C, et al. Aerobic and strength training in patients and care of patients with chronic obstructive pulmonary
with chronic obstructive pulmonary disease. Am J Respir disease. Am J Respir Crit Care Med 1995; 152:S77-121.
Crit Care Med 1999; 159:896-901. 150. Nocturnal Oxygen Therapy Trial Group. Continuous or
nocturnal oxygen therapy in hypoxemic chronic obstructive
lung disease: a clinical trial. Ann Intern Med 1980; 93:391-8.

84 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 85

151. Tarpy SP, Celli BR. Long-term oxygen therapy. N Engl J 168. Strumpf DA, Millman RP, Carlisle CC, Grattan LM, Ryan
Med 1995; 333:710-4. SM, Erickson AD, et al. Nocturnal positive-pressure ventila-
152. Report of the Medical Research Council Working Party. tion via nasal mask in patients with severe chronic obstruc-
Long term domiciliary oxygen therapy in chronic hypoxic tive pulmonary disease. Am Rev Respir Dis 1991;
cor pulmonale complicating chronic bronchitis and emphy- 144:1234-9.
sema. Lancet 1981; 1:681-6. 169. Casanova C, Celli BR, Tost L, Soriano E, Abreu J, Velasco
153. Weitzenblum E, Sautegeau A, Ehrhart M, Mammosser M, V, et al. Long-term controlled trial of nocturnal nasal posi-
Pelletier A. Long-term oxygen therapy can reverse the pro- tive pressure ventilation in patients with severe COPD.
gression of pulmonary hypertension in patients with chron- Chest 2000; 118:1582-90.
ic obstructive pulmonary disease. Am Rev Respir Dis 170. Clini E, Sturani C, Rossi A, Viaggi S, Corrado A, Donner
1985; 131:493-8. CF, et al. The Italian multicentre study on noninvasive ven-
154. Zielinski J, Tobiasz M, Hawrylkiewicz I, Sliwinski P, tilation in chronic obstructive pulmonary disease patients.
Palasiewicz G. Effects of long-term oxygen therapy on pul- Eur Respir J 2002; 20:529-38.
monary hemodynamics in COPD patients: a 6-year 171. Elliott MW. Noninvasive ventilation in chronic ventilatory
prospective study. Chest 1998; 113:65-70. failure due to chronic obstructive pulmonary disease. Eur
155. Petty TL. Supportive therapy in COPD. Chest 1998; Respir J 2002; 20:511-4.
113:256S-262S. 172. Consensus conference report. Clinical indications for non-
156. Petty TL, O'Donohue WJ, Jr. Further recommendations for invasive positive pressure ventilation in chronic respiratory
prescribing, reimbursement, technology development, and failure due to restrictive lung disease, COPD, and noctur-
research in long-term oxygen therapy. Summary of the nal hypoventilation. Chest 1999; 116:521-34.
Fourth Oxygen Consensus Conference, Washington, D.C., 173. Mehran RJ, Deslauriers J. Indications for surgery and
October 15-16, 1993. Am J Respir Crit Care Med 1994; patient work-up for bullectomy. Chest Surg Clin N Am
150:875-7. 1995; 5:717-34.
157. Pelletier-Fleury N, Lanoe JL, Fleury B, Fardeau M. The 174. Hughes JA, MacArthur AM, Hutchison DC, Hugh-Jones P.
cost of treating COPD patients with long-term oxygen ther- Long term changes in lung function after surgical treatment
apy in a French population. Chest 1996; 110:411-6. of bullous emphysema in smokers and ex-smokers. Thorax
158. Heaney LG, McAllister D, MacMahon J. Cost minimisation 1984; 39:140-2.
analysis of provision of oxygen at home: are the drug tariff 175. Laros CD, Gelissen HJ, Bergstein PG, Van den Bosch JM,
guidelines cost effective? BMJ 1999; 319:19-23. Vanderschueren RG, Westermann CJ, et al. Bullectomy for
159. Gong H, Jr. Air travel and oxygen therapy in cardiopul- giant bullae in emphysema. J Thorac Cardiovasc Surg
monary patients. Chest 1992; 101:1104-13. 1986; 91:63-70.
160. Berg BW, Dillard TA, Rajagopal KR, Mehm WJ. Oxygen 176. Cooper JD, Trulock EP, Triantafillou AN, Patterson GA, Pohl
supplementation during air travel in patients with chronic MS, Deloney PA, et al. Bilateral pneumectomy (volume
obstructive lung disease. Chest 1992; 101:638-41. reduction) for chronic obstructive pulmonary disease. J
161. Gong H, Jr., Tashkin DP, Lee EY, Simmons MS. Hypoxia- Thorac Cardiovasc Surg 1995; 109:106-16; discussion
altitude simulation test. Evaluation of patients with chronic 116-9.
airway obstruction. Am Rev Respir Dis 1984; 130:980-6. 177. Criner G, Cordova FC, Leyenson V, Roy B, Travaline J,
162. Christensen CC, Ryg M, Refvem OK, Skjonsberg OH. Sudarshan S, et al. Effect of lung volume reduction surgery
Development of severe hypoxaemia in chronic obstructive on diaphragm strength. Am J Respir Crit Care Med 1998;
pulmonary disease patients at 2,438 m (8,000 ft) altitude. 157:1578-85.
Eur Respir J 2000; 15:635-9. 178. Martinez FJ, de Oca MM, Whyte RI, Stetz J, Gay SE, Celli
163. Muir JF. Pulmonary rehabilitation in chronic respiratory BR. Lung-volume reduction improves dyspnea, dynamic
insufficiency. 5. Home mechanical ventilation. Thorax 1993; hyperinflation, and respiratory muscle function. Am J
48:1264-73. Respir Crit Care Med 1997; 155:1984-90.
164. Simonds AK. Negative pressure ventilation in acute hyper- 179. Fessler HE, Permutt S. Lung volume reduction surgery and
capnic chronic obstructive pulmonary disease. Thorax airflow limitation. Am J Respir Crit Care Med 1998;
1996; 51:1069-70. 157:715-22.
165. Corrado A, Gorini M, Villella G, De Paola E. Negative pres- 180. Cooper JD, Patterson GA, Sundaresan RS, Trulock EP,
sure ventilation in the treatment of acute respiratory failure: Yusen RD, Pohl MS, et al. Results of 150 consecutive bilat-
an old noninvasive technique reconsidered. Eur Respir J
eral lung volume reduction procedures in patients with
1996; 9:1531-44.
severe emphysema. J Thorac Cardiovasc Surg 1996;
166. Shapiro SH, Ernst P, Gray-Donald K, Martin JG, Wood-
112:1319-29; discussion 1329-30.
Dauphinee S, Beaupre A, et al. Effect of negative pressure
181. Russi EW, Stammberger U, Weder W. Lung volume reduc-
ventilation in severe chronic obstructive pulmonary dis-
tion surgery for emphysema. Eur Respir J 1997; 10:208-18.
ease. Lancet 1992; 340:1425-9.
182. Gelb AF, McKenna RJ, Jr., Brenner M, Schein MJ, Zamel
167. Hillberg RE, Johnson DC. Noninvasive ventilation. N Engl J
N, Fischel R. Lung function 4 years after lung volume
Med 1997; 337:1746-52.
reduction surgery for emphysema. Chest 1999; 116:1608-15.

MANAGEMENT OF COPD 85
GOLD_WR_05 8/18/05 12:56 PM Page 86

183. Brenner M, McKenna RJ, Jr., Gelb AF, Fischel RJ, Wilson 200. Monninkhof E, van der Valk P, van der Palen J, van
AF. Rate of FEV1 change following lung volume reduction Herwaarden C, Zielhuis G. Effects of a comprehensive self-
surgery. Chest 1998; 113:652-9. management programme in patients with chronic obstructive
184. National Emphysema Treatment Trial Research Group. pulmonary disease. Eur Respir J. 2003 Nov;22(5):815-20.
Patients at high risk of death after lung-volume-reduction 201. Datta D, Vitale A, Lahiri B, ZuWallack R. An evaluation of
surgery. N Engl J Med 2001; 345:1075-83. nebulized levalbuterol in stable COPD. Chest. 2003
185. Elpern EH, Behner KG, Klontz B, Warren WH, Szidon JP, Sep;124(3):844-9.
Kesten S. Lung volume reduction surgery: an analysis of 202. van der Valk P, Monninkhof E, van der Palen J, Zielhuis G,
hospital costs. Chest 1998; 113:896-9. van Herwaarden C. Effect of discontinuation of inhaled corti-
186. Albert RK, Lewis S, Wood D, Benditt JO. Economic aspects costeroids in patients with chronic obstructive pulmonary dis-
of lung volume reduction surgery. Chest 1996; 110:1068-71. ease: the COPE study. Am J Respir Crit Care Med
187. Geddes D, Davies M, Koyama H, Hansell D, Pastorino U, 2002;166(10):1358-63.
203. Hanania NA, Darken P, Horstman D, Reisner C, Lee B,
Pepper J, et al. Effect of lung-volume-reduction surgery in
Davis S, Shah T. The efficacy and safety of fluticasone pro-
patients with severe emphysema. N Engl J Med 2000; 343:239-45.
pionate(250microg)/salmeterol(50microg) combined in the
188. Benditt JO, Albert RK. Surgical options for patients with
advanced emphysema. Clin Chest Med 1997; 18:577-93. Diskus inhaler for the treatment of COPD. Chest. 2003
189. The National Emphysema Treatment Trial Research Group. Sep;124(3):834-43.
Rationale and design of The National Emphysema Treatment 204. Calverley PM, Boonsawat W, Cseke Z, Zhong N, Peterson S,
Trial: a prospective randomized trial of lung volume reduction Olsson H. Maintenance therapy with budesonide and for-
surgery. Chest 1999; 116:1750-61. moterol in chronic obstructive pulmonary disease. Eur Respir
190. Trulock EP. Lung transplantation. Am J Respir Crit Care Med J. 2003 Dec;22(6):912-9.
1997; 155:789-818. 205. Wongsurakiat P, Lertakyamanee J, Maranetra KN,
191. Theodore J, Lewiston N. Lung transplantation comes of age. Jongriratanakul S, Sangkaew S. Economic evaluation of
N Engl J Med 1990; 322:772-4. influenza vaccination in Thai chronic obstructive pulmonary
192. Hosenpud JD, Bennett LE, Keck BM, Fiol B, Boucek MM, disease patients. J Med Assoc Thai. 2003 Jun;86(6):497-508.
Novick RJ. The Registry of the International Society for 206. Honeyman P. Barr P. Stubbing DG. Effects of a walking aid on
Heart and Lung Transplantation: fifteenth official report-- disability, oxygenation, and breathlessness in patients with
1998. J Heart Lung Transplant 1998; 17:656-68. chronic airflow limitation. Journal of Cardiopulmonary
193. Annual report of the US scientific registry for transplant Rehabilitation. 16(1): 63-7, 1996
recipients and the Organ Procurement and Transplantation 207. Roomi J.Yohannes AM. Connolly MJ. The effect of walking aids
Network. Transplant data: 1988-1994. Washington, D.C.: on exercise capacity and oxygenation in elderly patients with
Division of Transplantation, Health Resources and Services chronic obstructive pulmonary disease. Age & Ageing.
Administration, US Department of Health and Human 27(6):703-6, 1998.
Services, 1995. 208. Yohannes AM, Connolly MJ. Early mobilization with walking aids
194. Hosenpud JD, Bennett LE, Keck BM, Edwards EB, Novick following hospital admission with acute exacerbation of chronic
RJ. Effect of diagnosis on survival benefit of lung transplan- obstructive pulmonary disease. Clin Rehabil. 2003 Aug;17(5):465-71.
tation for end- stage lung disease. Lancet 1998; 351:24-7. 209. Steiner MC, Barton RL, Singh SJ, Morgan MD. Nutritional
195. Maurer JR, Frost AE, Estenne M, Higenbottam T, Glanville enhancement of exercise performance in chronic obstructive
AR. International guidelines for the selection of lung trans- pulmonary disease: a randomised controlled trial. Thorax. 2003
plant candidates. The International Society for Heart and Sep;58(9):745-51.
Lung Transplantation, the American Thoracic Society, the 210. Nandi K, Smith AA, Crawford A, MacRae KD, Garrod R, Seed
American Society of Transplant Physicians, the European WA, Roberts CM. Oxygen supplementation before or after sub-
Respiratory Society. Transplantation 1998; 66:951-6. maximal exercise in patients with chronic obstructive pulmonary
196. Ramsey SD, Patrick DL, Albert RK, Larson EB, Wood DE, disease. Thorax. 2003 Aug;58(8):670-3.
Raghu G. The cost-effectiveness of lung transplantation. A 211. Lewis CA, Eaton TE, Young P, Kolbe J. Short-burst oxygen
pilot study. University of Washington Medical Center Lung immediately before and after exercise is ineffective in nonhypoxic
Transplant Study Group. Chest 1995; 108:1594-601. COPD patients. Eur Respir J. 2003 Oct;22(4):584-8.
197. Al MJ, Koopmanschap MA, van Enckevort PJ, Geertsma A, 212. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, Ries
van der Bij W, de Boer WJ, et al. Cost-effectiveness of lung A, Weinmann G, Wood DE; National Emphysema Treatment Trial
transplantation in The Netherlands: a scenario analysis. Research Group. A randomized trial comparing lung-volume-
Chest 1998; 113:124-30. reduction surgery with medical therapy for severe emphysema.
198. van Enckevort PJ, Koopmanschap MA, Tenvergert EM, N Engl J Med. 2003 May 2;348(21):2059-73.
Geertsma A, van der Bij W, de Boer WJ, et al. Lifetime costs 213. Goldstein RS, Todd TR, Guyatt G, Keshavjee S, Dolmage TE,
of lung transplantation: estimation of incremental costs. van Rooy S, Krip B, Maltais F, LeBlanc P, Pakhale S, Waddell
Health Econ 1997; 6:479-89. TK. Influence of lung volume reduction surgery (LVRS) on
199. van Enckevort PJ, TenVergert EM, Bonsel GJ, Geertsma A, health related quality of life in patients with chronic obstructive
van der Bij W, de Boer WJ, et al. Technology assessment of pulmonary disease. Thorax. 2003 May;58(5):405-10.
the Dutch Lung Transplantation Program. Int J Technol 214. Smetana GW. Preoperative pulmonary evaluation. N Engl J Med
Assess Health Care 1998; 14:344-56. 1999;340(12):937-44.

86 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 87

215. Trayner E, Jr., Celli BR. Postoperative pulmonary complications.


Med Clin North Am 2001;85(5):1129-39.
216. Weisman IM. Cardiopulmonary exercise testing in the preopera-
tive assessment for lung resection surgery. Semin Thorac
Cardiovasc Surg 2001;13(2):116-25.
217. Bolliger CT, Perruchoud AP. Functional evaluation of the lung
resection candidate. Eur Respir J 1998;11(1):198-212.
218. Schuurmans MM, Diacon AH, Bolliger CT. Functional evaluation
before lung resection. Clin Chest Med 2002;23(1):159-72.
219. Celli BR, MacNee W. Standards for the diagnosis and treatment
of patients with COPD: a summary of the ATS/ERS position
paper. Eur Respir J 2004;23(6):932-46.
220. Gallefoss F. The effects of patient education in COPD in a 1-
year follow-up randomised, controlled trial. Patient Educ Couns.
2004 Mar;52(3):259-66.
221. Man WD, Mustfa N, Nikoletou D, Kaul S, Hart N, Rafferty GF,
Donaldson N, Polkey MI, Moxham J. Effect of salmeterol on res-
piratory muscle activity during exercise in poorly reversible
COPD. Thorax. 2004 Jun;59(6):471-6.
222. O'Donnell DE, Fluge T, Gerken F, Hamilton A, Webb K, Aguilaniu
B, Make B, Magnussen H. Effects of tiotropium on lung hyperin-
flation, dyspnoea and exercise tolerance in COPD. Eur Respir J.
2004 Jun;23(6):832-40.
223. Oostenbrink JB, Rutten-van Molken MP, Al MJ, Van Noord JA,
Vincken W. One-year cost-effectiveness of tiotropium versus
ipratropium to treat chronic obstructive pulmonary disease. Eur
Respir J. 2004 Feb;23(2):241-9.
224. Spencer S, Calverley PM, Burge PS, Jones PW. Impact of pre-
venting exacerbations on deterioration of health status in COPD.
Eur Respir J. 2004 May;23(5):698-702.
225. Donohue JF, Kalberg C, Emmett A, Merchant K, Knobil K. A
short-term comparison of fluticasone propionate/ salmeterol with
ipratropium bromide/albuterol for the treatment of COPD. Treat
Respir Med. 2004;3(3):173-81.
226. Wongsurakiat P, Maranetra KN, Wasi C, Kositanont U,
Dejsomritrutai W, Charoenratanakul S. Acute respiratory illness
in patients with COPD and the effectiveness of influenza vacci-
nation: a randomized controlled study. Chest. 2004
Jun;125(6):2011-20.
227. Li J, Zheng JP, Yuan JP, Zeng GQ, Zhong NS, Lin CY. Protective
effect of a bacterial extract against acute exacerbation in patients
with chronic bronchitis accompanied by chronic obstructive pul-
monary disease. Chin Med J (Engl). 2004 Jun;117(6):828-34.

MANAGEMENT OF COPD 87
GOLD_WR_05 8/18/05 12:56 PM Page 88

COMPONENT 4: MANAGE EXACERBATIONS


The most common causes of an exacerbation are infection
KEY POINTS: of the tracheobronchial tree and air pollution10, but the
cause of about one-third of severe exacerbations cannot
be identified. The role of bacterial infections is controversial,
• Exacerbations of respiratory symptoms requiring med-
but recent investigations with newer research techniques
ical intervention are important clinical events in COPD.
have begun to provide important information.
Bronchoscopic studies have shown that at least 50% of
• The most common causes of an exacerbation are
patients have bacteria in high concentrations in their lower
infection of the tracheobronchial tree and air pollution, airways during exacerbations11. However, a significant
but the cause of about one-third of severe exacerba- proportion of these patients also have bacteria colonizing
tions cannot be identified (Evidence B). their lower airways in the stable phase of the disease.
There is some indication that the bacterial burden
• Inhaled bronchodilators (particularly inhaled ß2- increases during exacerbations11, and that acquisition
agonists and/or anticholinergics), theophylline, and of strains of the bacteria that are new to the patient is
systemic, preferably oral, glucocorticosteroids are associated with exacerbations12. Development of specific
effective treatments for exacerbations of COPD immune responses to the infecting bacterial strains, and
(Evidence A). the association of neutrophilic inflammation with bacterial
exacerbations also support the bacterial causation of a
• Patients experiencing COPD exacerbations with proportion of exacerbations13-16. Conditions that may
clinical signs of airway infection (e.g., increased mimic an exacerbation include pneumonia, congestive
volume and change of color of sputum, and/or fever) heart failure, pneumothorax, pleural effusion, pulmonary
may benefit from antibiotic treatment (Evidence B). embolism, and arrhythmia. Recommendations for use of
antibiotics for COPD exacerbations are provided at the
• Noninvasive intermittent positive pressure ventilation end of this chapter.
(NIPPV) in exacerbations improves blood gases
and pH, reduces in-hospital mortality, decreases the
need for invasive mechanical ventilation and DIAGNOSIS AND ASSESSMENT
intubation, and decreases the length of hospital
stay (Evidence A). OF SEVERITY
Medical History
Increased breathlessness, the main symptom of an
INTRODUCTION exacerbation, is often accompanied by wheezing and
chest tightness, increased cough and sputum, change
COPD is often associated with exacerbations of symptoms
of the color and/or tenacity of sputum, and fever.
1-4. In patients with Stage I: Mild COPD to Stage II:
Exacerbations may also be accompanied by a number
Moderate COPD, an exacerbation is associated with
of nonspecific complaints, such as malaise, insomnia,
increased breathlessness, often accompanied by
sleepiness, fatigue, depression, and confusion. A
increased cough and sputum production, and may
decrease in exercise tolerance, fever, and/or new radio-
require medical attention outside of the hospital5. The
logical anomalies suggestive of pulmonary disease may
need for medical intervention intensifies as the airflow
herald a COPD exacerbation. An increase in sputum
limitation worsens. Exacerbations in Stage IV: Very
volume and purulence points to a bacterial cause, as
Severe COPD are associated with acute respiratory failure,
does prior history of chronic sputum production4,14.
representing a significant burden on the health care
system. Hospital mortality of patients admitted for an
exacerbation of COPD is approximately 10%, and the
Assessment of Severity
long-term outcome is poor. Mortality reaches 40% in one
Assessment of the severity of an exacerbation is based
year6-9, and is even higher (up to 59%) for patients older
on the patient’s medical history before the exacerbation,
than 65 years9. These figures vary from country to country
symptoms, physical examination, lung function tests,
depending on the health care system and the availability
arterial blood gas measurements, and other laboratory
of intensive care unit (ICU) beds.
tests (Figure 5-4-1). Specific information is required on

88 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 89

the frequency and severity of attacks of breathlessness Chest X-ray and ECG. Chest radiographs
and cough, sputum volume and color, and limitation (posterior/anterior plus lateral) are useful in identifying
of daily activities. When available, prior tests of lung alternative diagnoses that can mimic the symptoms of an
function and arterial blood gases are extremely useful for exacerbation. Although the history and physical signs
comparison with those made during the acute episode, can be confusing, especially when pulmonary hyperinflation
as an acute change in these tests is more important than masks coexisting cardiac signs, most problems are
their absolute values. Thus, where possible, physicians resolved by the chest X-ray and ECG. An ECG aids in
should instruct their patients to bring the summary of the diagnosis of right heart hypertrophy, arrhythmias, and
their last evaluation when they come to the hospital with ischemic episodes. Pulmonary embolism can be very
an exacerbation. In patients with very severe COPD, the difficult to distinguish from an exacerbation, especially in
most important sign of a severe exacerbation is a change severe COPD, because right ventricular hypertrophy and
in the alertness of the patient and this signals a need for large pulmonary arteries lead to confusing ECG and
immediate evaluation in the hospital. radiographic results. Spiral CT scanning and angiogra-
phy, and perhaps specific D-dimer assays, are the best
Lung function tests. Even simple lung function tests tools presently available for the diagnosis of pulmonary
can be difficult for a sick patient to perform properly. In embolism in patients with COPD, but ventilation-perfusion
general, a PEF < 100 L/min or an FEV1 < 1.00 L scanning is of no value. A low systolic blood pressure
indicates a severe exacerbation21-23, except in patients and an inability to increase the PaO2 above 8.0 kPa
with chronically severe airflow limitation. For instance, (60 mm Hg) despite high-flow oxygen also suggest
an FEV1 of 0.75 L, or a PaO2/FiO2 (FiO2 = fractional pulmonary embolism. If there are strong indications that
concentration of oxygen in dry inspired gas) of 32 kPa pulmonary embolism has occurred, it is best to treat for
(240 mm Hg) may be well tolerated by a subject with this along with the exacerbation.
severe COPD who copes with these values in stable
conditions, whereas they may reflect a severe Other laboratory tests. The whole blood count may
exacerbation for a subject with slightly higher values, e.g., identify polycythemia (hematocrit > 55%) or bleeding.
an FEV1 of 0.90 L or a PaO2/FiO2 of 38 kPa (282 mm White blood cell counts are usually not very informative.
Hg) in stable conditions24. The presence of purulent sputum during an exacerbation
of symptoms is sufficient indication for starting empirical
antibiotic treatment. Streptococcus pneumoniae,
Figure 5-4-1. Medical History and Signs of Hemophilis influenzae, and Moraxella catarrhalis are the
Severity of Exacerbations of COPD most common bacterial pathogens involved in COPD
exacerbations. If an infectious exacerbation does not
Medical History Signs of Severity respond to the initial antibiotic treatment, a sputum
culture and an antibiogram should be performed.
• Duration of worsening or • Use of accessory respira-
Biochemical tests can reveal whether the cause of
new symptoms. tory muscles.
the exacerbation is an electrolyte disturbance(s)
• Number of previous • Paradoxical chest wall (hyponatremia, hypokalemia, etc.), a diabetic crisis, or
episodes movements. poor nutrition (low proteins), and may suggest a
(exacerbations/hospital- • Worsening or new onset metabolic acid-base disorder.
izations). central cyanosis.
• Present treatment regimen. • Development of peripheral HOME MANAGEMENT
edema.
There is increasing interest in home care for end-stage
• Hemodynamic instability.
COPD patients, although economic studies of home-care
• Signs of right heart failure. services have yielded mixed results. Four randomized
• Reduced alertness. clinical trials have shown nurse administered home care
represents an effective and practical alternative to
hospitalization in selected patients with exacerbations of
Arterial blood gases. In the hospital, measurement of
COPD without acidotic respiratory failure. However, the
arterial blood gases is essential to assess the severity of
exact criteria for home vs hospital treatment remains
an exacerbation. A PaO2 < 8.0 kPa (60 mm Hg) and/or
uncertain and will vary by health care setting26-29. A major
SaO2 < 90% with or without PaCO2 > 6.7 kPa, (50 mm Hg)
outstanding issue is when to treat an exacerbation at
when breathing room air indicate respiratory failure. In
home and when to hospitalize the patient.
addition, a PaO2 < 6.7 kPa (50 mm Hg), PaCO2 > 9.3
kPa (70 mm Hg), and pH < 7.30 point toward a life-threat-
ening episode that needs critical management25.

MANAGEMENT OF COPD 89
GOLD_WR_05 8/18/05 12:56 PM Page 90

Figure 5-4-2. Algorithm for the Management HOSPITAL MANAGEMENT


of an Exacerbation of COPD at Home
The risk of dying from an exacerbation of COPD is
Initiate or increase bronchodilator therapy closely related to the development of respiratory acidosis,
Consider antibiotics the presence of significant comorbidities, and the need
for ventilatory support6. Patients lacking these features
Reassess within hours
are not at high risk of dying, but those with severe
underlying COPD often require hospitalization in any case.
Attempts at managing such patients entirely in the
Resolution or improvement No resolution or improvement community have met with only limited success38, but
of signs and symptoms returning them to their homes with increased social
support and a supervised medical care package after
Add oral corticosteroids initial emergency room assessment has been much more
successful39. Several randomized controlled trials have
Continue management
Step down when possible confirmed that this is a safe alternative to hospitalization,
Reassess within hours
although it probably only applies to about 25% of COPD
admissions. Savings on inpatient expenditures40 offset the
Worsening of signs/symptoms
additional costs of maintaining a community-based
Review long-term management
COPD nursing team. However, detailed cost-benefit
analyses of these approaches are awaited.
Refer to hospital
Figure 5-4-3. Indications for Hospital Assessment
or Admission for Exacerbations of COPD*
The algorithm reported in Figure 5-4-2 may assist in
the management of an exacerbation at home; a stepwise
• Marked increase in intensity of symptoms,
therapeutic approach is recommended30-33.
such as sudden development of resting dyspnea.
Bronchodilator Therapy • Severe background COPD.
• Onset of new physical signs (e.g., cyanosis,
Home management of COPD exacerbations involves
increasing the dose and/or frequency of existing peripheral edema).
bronchodilator therapy (Evidence A). If not already • Failure of exacerbation to respond to initial
used, an anticholinergic can be added until the symptoms medical management.
improve. In more severe cases, high-dose nebulizer
• Significant comorbidities.
therapy can be given on an as-needed basis for several
days and if a suitable nebulizer is available. However, • Newly occurring arrhythmias.
long-term use of nebulizer therapy after an acute episode • Diagnostic uncertainty.
is not routinely recommended.
• Older age.
Glucocorticosteroids • Insufficient home support.

Systemic glucocorticosteroids are beneficial in the Figure 5-4-4. Indications for ICU Admission of
management of exacerbations of COPD. They shorten Patients with Exacerbations of COPD*
recovery time and help to restore lung function more
quickly34-36 (Evidence A) and may reduce the risk of early • Severe dyspnea that responds inadequately to
relapse73. They should be considered in addition to initial emergency therapy.
bronchodilators if the patient’s baseline FEV1 is < 50%
predicted. A dose of 40 mg prednisolone per day for 10 • Confusion, lethargy, coma.
days is recommended (Evidence D). One large study • Persistent or worsening hypoxemia (PaO2
indicates that nebulized budesonide may be an alternative < 5.3 kPa, 40 mm Hg), and/or severe/worsening
to oral glucocorticosteroids in the treatment hypercapnia (PaCO2 > 8.0 kPa, 60 mm Hg), and/or
of nonacidotic exacerbations37. severe/worsening respiratory acidosis (pH < 7.25)
despite supplemental oxygen and NIPPV.

*Local resources need to be considered.

90 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 91

Controlled oxygen therapy. Oxygen therapy is the


Figure 5-4-5. Management of Severe but cornerstone of hospital treatment of COPD exacerba-
Not Life-Threatening Exacerbations of COPD in the tions. Adequate levels of oxygenation (PaO2 > 8.0 kPa,
60 mm Hg, or SaO2 > 90%) are easy to achieve in
Emergency Department or the Hospital*
uncomplicated exacerbations, but CO2 retention can
occur insidiously with little change in symptoms. Once
• Assess severity of symptoms, blood gases, chest oxygen is started, arterial blood gases should be checked
X-ray.
30 minutes later to ensure satisfactory oxygenation
• Administer controlled oxygen therapy and repeat without CO2 retention or acidosis. Venturi masks are
arterial blood gas measurement after 30 minutes. more accurate sources of controlled oxygen than are
• Bronchodilators: nasal prongs but are more likely to be removed by the
patient.
– Increase doses or frequency.
– Combine ß2-agonists and anticholinergics. Bronchodilator therapy. Short-acting inhaled
2-agonists are usually the preferred bronchodilators for
– Use spacers or air-driven nebulizers. treatment of exacerbations of COPD30,31,41 (Evidence A). If
– Consider adding intravenous aminophylline, if a prompt response to these drugs does not occur, the
needed. addition of an anticholinergic is recommended, even
though evidence concerning the effectiveness of this
• Add oral or intravenous glucocorticosteroids.
combination is controversial. Despite its wide-spread
• Consider antibiotics: clinical use, the role of aminophylline in the treatment of
– When signs of bacterial infection, oral or exacerbations of COPD remains controversial. Most
occasionally intravenous. studies of aminophylline have demonstrated minor
improvements in lung volumes without showing gas
• Consider noninvasive mechanical ventilation. exchange deterioration42,43. In more severe exacerbations,
• At all times: addition of an oral or intravenous methylxanthine to the
treatment can be considered. However, close monitoring
– Monitor fluid balance and nutrition.
of serum theophylline is recommended to avoid the side
– Consider subcutaneous heparin. effects of these drugs42,44-46. Possible beneficial effects in
– Identify and treat associated conditions lung function, and clinical endpoints, are modest and
(e.g., heart failure, arrhythmias). inconsistent, whereas adverse effects are significantly
increased74.
– Closely monitor condition of the patient.
Glucocorticosteroids. Oral or intravenous glucocorti-
*Local resources need to be considered. costeroids are recommended as an addition to
bronchodilator therapy (plus eventually antibiotics
A range of criteria to consider for hospital assessment/ and oxygen therapy) in the hospital management of
admission for exacerbations of COPD are shown in exacerbations of COPD35-36 (Evidence A). The exact
Figure 5-4-3. Some patients need immediate admission dose that should be recommended is not known, but high
to an intensive care unit (ICU) (Figure 5-4-4). Admission doses are associated with a significant risk of side effects.
of patients with severe COPD exacerbations to intermediate Thirty to 40 mg of oral prednisolone daily for 10 to 14
or special respiratory care units may be appropriate if
days is a reasonable compromise between efficacy and
personnel, skills, and equipment exist to identify and
safety (Evidence D). Prolonged treatment does not result
manage acute respiratory failure successfully.
in greater efficacy and increases the risk of side effects.
Emergency Department or Hospital Ventilatory support. The primary objectives of
mechanical support in patients with exacerbations in
The first actions when a patient reaches the emergency
Stage IV: Very Severe COPD are to decrease mortality
department are to provide controlled oxygen therapy
and morbidity and to relieve symptoms. Ventilatory
and to determine whether the exacerbation is life
support includes both noninvasive mechanical ventilation
threatening. If so, the patient should be admitted to the
using either negative or positive pressure devices, and
ICU immediately. Otherwise, the patient may be
invasive (conventional) mechanical ventilation by
managed in the emergency department or hospital as
oro/naso-tracheal tube or tracheostomy.
detailed in Figure 5-4-5.

MANAGEMENT OF COPD 91
GOLD_WR_05 8/18/05 12:56 PM Page 92

Figure 5-4-6. Indications and Relative Figure 5-4-7. Indications for


Contraindications for NIPPV47,56 Invasive Mechanical Ventilation

• Severe dyspnea with use of accessory muscles


Selection criteria
and paradoxical abdominal motion.
• Moderate to severe dyspnea with use of accessory
• Respiratory frequency > 35 breaths per minute.
muscles and paradoxical abdominal motion.
• Life-threatening hypoxemia (PaO2 < 5.3 kPa,
• Moderate to severe acidosis (pH ≤ 7.35) and
40 mm Hg or PaO2/FiO2 < 200 mm Hg).
hypercapnia (PaCO2 > 6.0 kPa, 45 mm Hg)57.
• Severe acidosis (pH < 7.25) and hypercapnia
• Respiratory frequency > 25 breaths per minute.
(PaCO2 > 8.0 kPa, 60 mm Hg).
• Respiratory arrest.
Exclusion criteria (any may be present)
• Respiratory arrest. • Somnolence, impaired mental status.
• Cardiovascular instability (hypotension, • Cardiovascular complications (hypotension,
shock, heart failure).
arrhythmias, myocardial infarction).
• Somnolence, impaired mental status, • Other complications (metabolic abnormalities,
sepsis, pneumonia, pulmonary embolism,
uncooperative patient.
barotrauma, massive pleural effusion).
• High aspiration risk; viscous or copious secretions.
• NIPPV failure (or exclusion criteria,
• Recent facial or gastroesophageal surgery.
see Figure 5-4-7).
• Craniofacial trauma, fixed nasopharyngeal
abnormalities.
FiO2: Fractional concentration of oxygen in dry inspired gas.
• Burns.
• Extreme obesity.
Figure 5-4-8. Factors Determining
Benefit from Invasive Ventilation
Noninvasive mechanical ventilation. Noninvasive
intermittent positive pressure ventilation (NIPPV) has
• Cultural attitudes toward chronic disability.
been studied in many uncontrolled and five randomized
controlled trials in acute respiratory failure47,48. The
• Expectations of therapy.
studies show consistently positive results with success • Financial resources (especially the provision
rates of 80-85%49. Taken together they provide evidence of ICU facilities).
that NIPPV increases pH, reduces PaCO2, reduces • Perceived likelihood of recovery.
the severity of breathlessness in the first 4 hours of • Customary medical practice.
treatment, and decreases the length of hospital stay • Wishes, if known, of the patient.
(Evidence A). More importantly, mortality - or its surrogate,
intubation rate - is reduced by this intervention50-53.
However, NIPPV is not appropriate for all patients, as life-threatening acid-base status abnormalities and/or
summarized in Figure 5-4-649. altered mental status despite aggressive pharmacologic
therapy are likely to be the best candidates for invasive
Invasive (conventional) mechanical ventilation. During (conventional) mechanical ventilation. The indications for
exacerbations of COPD the events occurring within the initiating mechanical ventilation during exacerbations of
lungs include bronchoconstriction, airway inflammation, COPD are shown in Figure 5-4-7, the first being the
increased mucous secretions, and loss of elastic recoil, commonest and most important reason. Figure 5-4-8
all of which prevent the respiratory system from reaching details the factors determining benefit from invasive
its passive functional residual capacity at the end of ventilation. The three ventilatory modes most widely
expiration, enhancing dynamic hyperinflation54. As a used are assisted-control ventilation, and pressure
result of these processes, an elastic threshold load, support ventilation alone or in combination with intermittent
referred to as intrinsic or auto-positive end-expiratory mandatory ventilation55.
pressure (PEEPi), is imposed on the inspiratory muscles
at the beginning of inspiration and increases the work of The use of invasive ventilation in end-stage COPD
breathing. For these reasons, patients who show patients is influenced by the likely reversibility of the
impending acute respiratory failure and those with precipitating event, the patient’s wishes, and the availability

92 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 93

of intensive care facilities. Major hazards include the risk ment). Manual or mechanical chest percussion and
of ventilator-acquired pneumonia (especially when postural drainage may be beneficial in patients producing
multi-resistant organisms are prevalent), barotrauma, > 25 ml sputum per day or with lobar atelectasis.
and failure to wean to spontaneous ventilation. Contrary
to some opinions, mortality among COPD patients with Hospital Discharge and Follow-Up
respiratory failure is no greater than mortality among
patients ventilated for non-COPD causes. Insufficient clinical data exist to establish the optimal
duration of hospitalization in individual patients
A review of a large number of North American COPD developing an exacerbation of COPD1,66,67. Consensus
patients ventilated for respiratory failure indicated an in and limited data support the discharge criteria listed in
hospital mortality of 17-30%58. Further attrition over the Figure 5-4-9. Figure 5-4-10 provides items to include in
next 12 months was particularly high among those a follow-up assessment 4 to 6 weeks after discharge from
patients who had poor lung function before ventilation the hospital. Thereafter, follow-up is the same as for
(FEV1 < 30% predicted), had a non-respiratory comorbidity, stable COPD, including supervising smoking cessation,
or were housebound. Patients who did not have a monitoring the effectiveness of each drug treatment, and
previously diagnosed underlying disease, had respiratory monitoring changes in spirometric parameters39. Home
failure due to a potentially reversible cause (such as an visits by a community nurse may permit earlier discharge
infection), or were relatively mobile and not using long- of patients hospitalized with an exacerbation of COPD,
term oxygen did surprisingly well with ventilatory support. without increasing readmission rate29,68-70. Early outpatient
When possible, a clear statement of the patient’s own pulmonary rehabilitation after hospitalization for COPD
treatment wishes - an advance directive or “living will” - exacerbation results in exercise capacity and health
makes these difficult decisions much easier to resolve. status improvements at three months75.

Weaning or discontinuation from mechanical ventilation If hypoxemia developed during the exacerbation, arterial
can be particularly difficult and hazardous in patients with blood gases should be rechecked at discharge and at
COPD. The most influential determinant of mechanical the follow-up visit. If the patient remains hypoxemic,
ventilatory dependency in these patients is the balance long-term oxygen therapy should be instituted. Decisions
between the respiratory load and the capacity of the about suitability for continuous domiciliary oxygen based
respiratory muscles to cope with this load59. By contrast, on the severity of the acute hypoxemia during an
pulmonary gas exchange by itself is not a major difficulty exacerbation are frequently misleading.
in patients with COPD60-62. Weaning patients from the
ventilator can be a very difficult and prolonged process The opportunities for prevention of future exacerbations
and the best method remains a matter of debate63,64. should be reviewed before discharge, with particular
Whether pressure support or a T-piece trial is used,
weaning is shortened when a clinical protocol is adopted
(Evidence A). Noninvasive ventilation has been applied Figure 5-4-9. Discharge Criteria for
to facilitate the weaning process in COPD patients with Patients with Exacerbations of COPD
acute or chronic respiratory failure58. Compared with
invasive pressure support ventilation, noninvasive • Inhaled ß2-agonist therapy is required no more
intermittent positive pressure ventilation (NIPPV) during frequently than every 4 hrs.
weaning shortened weaning time, reduced the stay in • Patient, if previously ambulatory, is able to walk
the intensive care unit, decreased the incidence of across room.
nosocomial pneumonia, and improved 60-day survival
rates. Similar findings have been reported when NIPPV is • Patient is able to eat and sleep without frequent
used after extubation for hypercapnic respiratory failure65 awakening by dyspnea.
(Evidence C). • Patient has been clinically stable for 12-24 hrs.
• Arterial blood gases have been stable for 12-24 hrs.
Other measures. Further treatments that can be used in
• Patient (or home caregiver) fully understands correct
the hospital include: fluid administration (accurate monitoring
use of medications.
of fluid balance is essential); nutrition (supplementary
when the patient is too dyspneic to eat); low molecular • Follow-up and home care arrangements have been
heparin in immobilized, polycythemic, or dehydrated completed (e.g., visiting nurse, oxygen delivery, meal
patients with or without a history of thromboembolic provisions).
disease; and sputum clearance (by stimulating coughing • Patient, family, and physician are confident patient
and low-volume forced expirations as in home manage- can manage successfully.

MANAGEMENT OF COPD 93
GOLD_WR_05 8/18/05 12:56 PM Page 94

• Patients with exacerbations of COPD with two of the


Figure 5-4-10. Follow-Up Assessment 4-6 Weeks cardinal symptoms, if increased purulence of sputum
After Discharge from Hospital for is one of the two symptoms. (Evidence C)
Exacerbations of COPD
• Patients with a severe exacerbation of COPD that
requires invasive mechanical ventilation (invasive and
• Ability to cope in usual environment. non-invasive). (Evidence B)
• Measurement of FEV1.
• Reassessment of inhaler technique. The predominant bacterial organisms recovered in the
• Understanding of recommended treatment regimen. lower airways of patients with mild exacerbations are
• Need for long-term oxygen therapy and/or home H. influenzae, S. pneumoniae and M. catarrhalis11,80.
nebulizer (for patients with very severe COPD). In contrast, studies in patients requiring mechanical
ventilation with severe underlying COPD81,82 have shown
that other microorganisms, such as enteric gram negative
attention to future influenza vaccination plans, knowledge bacilli and P. aeruginosa may be more frequent. Other
about current therapy including inhaler technique71,72, and studies have shown that the severity of the COPD is an
how to recognize symptoms of exacerbations. important determinant of the type of microorganism83,84.
In patients with mild COPD, S. pneumoniae is predominant.
Pharmacotherapy known to reduce the number of When the FEV1 is lower, H. influenzae and M. catarrhalis
exacerbations should be considered. Social problems are more frequent and P. aeruginosa may appear in
should be discussed and principal caregivers identified if patients with a more severe degree of airways obstruction
the patient has a significant persisting disability. (Figure 5-4-11). The risk factors for P. aeruginosa infection
are recent hospitalisation, frequent administration of antibiotics
Antibiotics (4 courses in the last year, very severe COPD (Stage IV),
and isolation of P. aeruginosa during a previous
Randomised placebo controlled studies of antibiotic exacerbation or colonization during a stable period83,84.
treatment in exacerbations of COPD have demonstrated
a small beneficial effect of antibiotics on lung function76, There is no clear information about when to use oral or
and one randomised controlled trial has provided evidence intravenous route of administration in hospitalized
for a significant beneficial effect of antibiotics in COPD patients. The route of administration depends on the
patients who presented with an increase in all three of the ability of the patient to eat, and the pharmacokinetics of
following cardinal symptoms: dyspnea, sputum volume, the antibiotic. The oral route is preferred. Otherwise,
sputum purulence77. There was also some benefit in the IV route has to be used, switching to oral when there
those patients with an increase in only two of these cardinal is clinical stabilization. Antibiotic treatment in patients
symptoms. with exacerbations of COPD should be maintained for 3
to 10 days. Figure 5-4-12 provides recommended
A study on non-hospitalized patients with exacerbations antibiotic treatment in exacerbations of COPD.
of COPD showed a relationship between the purulence of
the sputum and the presence of bacteria78, suggesting Ten to thirty percent of COPD exacerbated patients do
that these patients should be treated with antibiotics if not respond to empiric antimicrobial treatment80. In such
they also have at least one of the other two cardinal cases the patient should be re-evaluated for complications
symptoms (dyspnea or sputum volume). However, that can aggravate symptoms and mimic exacerbations
these criteria for exacerbations of COPD have not been (e.g., cardiac failure, pulmonary embolism, non-compliance
validated in other studies. A study in COPD patients with prescribed medications); microbiological reassessment
with exacerbations requiring mechanical ventilation of these patients is recommended.
(invasive and non-invasive) indicated that not giving
antibiotics was associated with increased mortality and
a greater incidence of secondary intra-hospital pneumonia79.

Based on the current available evidence, antibiotics


should be given to:

• Patients with exacerbations of COPD with three of


the following cardinal symptoms: increased dyspnea,
increased sputum volume, increased sputum purulence.
(Evidence B)

94 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 95

Figure 5-4-11: Stratification of patients with COPD exacerbated for antibiotic


treatment and potential microorganisms involved in each group
Groupa Definitionb Microorganisms
Group A: Patients not requiring Mild exacerbation H. influenzae
hospitalization (Stage I: Mild COPD) S. pneumoniae
M. catarrhalis
Chlamydia pneumoniae c
Viruses
Group B: Patients admitted to hospital Moderate-severe exacerbation without Group A plus:
(Stages II-IV: Moderate to Very Severe risk factors for P. aeruginosa infection Enterobacteriaceae (K.pneumoniae,
COPD) E. coli, Proteus, Enterobacter, etc)
Group C: Patients admitted to hospital Moderate-severe exacerbation with risk Group B plus:
(Stages II-IV: Moderate to Very Severe factors for P. aeruginosa infection P. aeruginosa
COPD)
a. In some settings, patients with moderate to severe exacerbations may be treated as outpatients. In this case, patients may best be stratified into
two groups: an uncomplicated group without any risk factors and a complicated group that has one or more ‘risk factors’ (co-morbidity, severe COPD,
frequent exacerbations, antimicrobial use within last 3 months). The uncomplicated group: use Group A recommendations Figure 5-4-12.
Complicated group: use Group B or C recommendations (oral therapy) Figure 5-4-12217-19.
b. Severity refers to the exacerbation, though this is intertwined with the severity of the underlying COPD.
c. Chlamydia pneumonia (or Chlamidophila pneumoniae) has not been confirmed as a cause of exacerbations in some areas (e.g., UK).

Figure 5-4-12: Antibiotic treatment in exacerbations of COPD a,b


Oral Treatment Alternative Parental Treatment
(No particular order) (No particular order) (No particular order)

Group A Patients with only one cardinal symptom


should not receive antibiotics
If indication then: • ß-lactam/ß-lactamase inhibitor
• ß-lactam (Ampicillin/Amoxicillinc) (Co-amoxiclav)

• Tetracycline • Macrolides (Azithromycin,


Clarithromycin, Roxithromycind)
• Trimethoprim/Sulfamethoxazole
• Cephalosporins - 2nd or 3rd generation
• Ketolides (Telithromycin)
Group B • ß-lactam/b-lactamase inhibitor • Fluoroquinolonesd (Gatifloxacin, • ß-lactam/b-lactamase inhibitor
(Co-amoxiclav) Gemifloxacin, Levofloxacin, (Co-amoxiclav, ampicillin/sulbactam)
Moxifloxacin)
• Cephalosporins - 2nd or
3rd generation
• Fluoroquinolonesd (Gatifloxacin,
Levofloxacin, Moxifloxacin)
Group C • Fluoroquinolones (Ciprofloxacin, • Fluoroquinolones (Ciprofloxacin,
Levofloxacin - high dosee) Levofloxacin - high dosee) or
• ß-lactam with P.aeruginosa activity

a. All patients with symptoms of a COPD exacerbation should be treated with additional bronchodilators ± glucocorticosteroids.
b. Classes of antibiotics are provided (with specific agents in parentheses). In countries with high incidence of S. pneumoniae resistant to penicillin,
high dosages of Amoxicillin or Co-Amoxiclav are recommended. (See Table 1 for definition of Groups A, B, C.)
c. This antibiotic is not appropriate in areas where there is increased prevalence of ß-lactamase producing H. influenzae and M. catarrhalis and/or of
S. pneumoniae resistant to penicillin.
d. Not available in all areas of the world.
e. Dose 750 mgs effective against P. aeruginosa.

MANAGEMENT OF COPD 95
GOLD_WR_05 8/18/05 12:56 PM Page 96

REFERENCES 14. Murphy TF, Brauer AL, Grant BJ, Sethi S. Moraxella
catarrhalis in Chronic Obstructive Pulmonary Disease:
1. Regueiro CR, Hamel MB, Davis RB, Desbiens N, Connors Burden of Disease and Immune Response. Am J Respir
AF, Jr., Phillips RS. A comparison of generalist and pulmo- Crit Care Med 2005; 172(2):195-9.
nologist care for patients hospitalized with severe chronic 15. Sethi S, Muscarella K, Evans N, Klingman KL, Grant BJ,
obstructive pulmonary disease: resource intensity, hospital Murphy TF. Airway inflammation and etiology of acute
costs, and survival. SUPPORT Investigators. Study to exacerbations of chronic bronchitis. Chest 2000;
Understand Prognoses and Preferences for Outcomes and 118(6):1557-65.
Risks of Treatment. Am J Med 1998; 105:366-72. 16. White AJ, Gompertz S, Bayley DL, Hill SL, O'Brien C,
2. Gibson PG, Wlodarczyk JH, Wilson AJ, Sprogis A. Severe Unsal I, Stockley RA. Resolution of bronchial inflammation
exacerbation of chronic obstructive airways disease: health is related to bacterial eradication following treatment of
resource use in general practice and hospital. J Qual Clin exacerbations of chronic bronchitis. Thorax 2003; 58(8):680-5.
Pract 1998; 18:125-33. References 17 - 20 have been deleted.
3. Warren PM, Flenley DC, Millar JS, Avery A. Respiratory 21. Emerman CL, Effron D, Lukens TW. Spirometric criteria for
failure revisited: acute exacerbations of chronic bronchitis hospital admission of patients with acute exacerbation of
between 1961-68 and 1970-76. Lancet 1980; 1:467-70. COPD. Chest 1991; 99:595-9.
4. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES, 22. Emerman CL, Lukens TW, Effron D. Physician estimation
Harding GK, Nelson NA. Antibiotic therapy in exacerba- of FEV1 in acute exacerbation of COPD. Chest 1994;
tions of chronic obstructive pulmonary disease. Ann Intern 105:1709-12.
Med 1987; 106:196-204. 23. Emerman CL, Cydulka RK. Use of peak expiratory flow
5. Thompson AB, Mueller MB, Heires AJ, Bohling TL, rate in emergency department evaluation of acute exacer-
Daughton D, Yancey SW, et al. Aerosolized beclometha- bation of chronic obstructive pulmonary disease. Ann
sone in chronic bronchitis. Improved pulmonary function Emerg Med 1996; 27:159-63.
and diminished airway inflammation. Am Rev Respir Dis 24. Barbera JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N,
1992; 146:389-95. et al. Mechanisms of worsening gas exchange during
6. Connors AF, Jr., Dawson NV, Thomas C, Harrell FE, Jr., acute exacerbations of chronic obstructive pulmonary dis-
Desbiens N, Fulkerson WJ, et al. Outcomes following acute ease. Eur Respir J 1997; 10:1285-91.
exacerbation of severe chronic obstructive lung disease. 25. Emerman CL, Connors AF, Lukens TW, Effron D, May ME.
The SUPPORT investigators (Study to Understand Relationship between arterial blood gases and spirometry
Prognoses and Preferences for Outcomes and Risks of in acute exacerbations of chronic obstructive pulmonary
Treatments). Am J Respir Crit Care Med 1996; 154:959-67. disease. Ann Emerg Med 1989; 18:523-7.
7. Kong GK, Belman MJ, Weingarten S. Reducing length of 26. Davies L, Wilkinson M, Bonner S, Calverley PM, Angus
stay for patients hospitalized with exacerbation of COPD RM. "Hospital at home" versus hospital care in patients
by using a practice guideline. Chest 1997; 111:89-94. with exacerbations of chronic obstructive pulmonary dis-
8. Fuso L, Incalzi RA, Pistelli R, Muzzolon R, Valente S, ease: prospective randomised controlled trial. BMJ 2000;
Pagliari G, et al. Predicting mortality of patients hospital- 321:1265-8.
ized for acutely exacerbated chronic obstructive pulmonary 27. Ojoo JC, Moon T, McGlone S, Martin K, Gardiner ED,
disease. Am J Med 1995; 98:272-7. Greenstone MA, et al. Patients' and carers' preferences in
9. Seneff MG, Wagner DP, Wagner RP, Zimmerman JE, two models of care for acute exacerbations of COPD:
Knaus WA. Hospital and 1-year survival of patients admitted results of a randomised controlled trial. Thorax 2002;
to intensive care units with acute exacerbation of chronic 57:167-9.
obstructive pulmonary disease. JAMA 1995; 274:1852-7. 28. Skwarska E, Cohen G, Skwarski KM, Lamb C, Bushell D,
10. White AJ, Gompertz S, Stockley RA. Chronic obstructive Parker S, et al. Randomized controlled trial of supported
pulmonary disease . 6: The aetiology of exacerbations of discharge in patients with exacerbations of chronic
chronic obstructive pulmonary disease. Thorax 2003; obstructive pulmonary disease. Thorax 2000; 55:907-12.
58(1):73-80. 29. Hernandez C, Casas A, Escarrabill J, Alonso J, Puig-Junoy
11. Monso E, Ruiz J, Rosell A, Manterola J, Fiz J, Morera J, J, Farrero E, et al. Home hospitalisation of exacerbated
Ausina V. Bacterial infection in chronic obstructive pul- chronic obstructive pulmonary disease patients. Eur Respir
monary disease. A study of stable and exacerbated outpa- J 2003; 21:58-67.
tients using the protected specimen brush. Am J Respir 30. Siafakas NM, Vermeire P, Pride NB, Paoletti P, Gibson J,
Crit Care Med 1995; 152(4 Pt 1):1316-20. Howard P, et al. Optimal assessment and management of
12. Sethi S, Evans N, Grant BJ, Murphy TF. New strains of chronic obstructive pulmonary disease (COPD). The
bacteria and exacerbations of chronic obstructive pul- European Respiratory Society Task Force. Eur Respir J
monary disease. N Engl J Med 2002; 347(7):465-71. 1995; 8:1398-420.
13. Sethi S, Wrona C, Grant BJ, Murphy TF. Strain-specific 31. American Thoracic Society. Standards for the diagnosis
immune response to Haemophilus influenzae in chronic and care of patients with chronic obstructive pulmonary
obstructive pulmonary disease. Am J Respir Crit Care Med disease. Am J Respir Crit Care Med 1995; 152:S77-121.
2004; 169(4):448-53. 32. Sachs FL. Chronic bronchitis. Clin Chest Med 1981; 2:79-89.

96 MANAGEMENT OF COPD
GOLD_WR_05 8/18/05 12:56 PM Page 97

33. Celli BR. Current thoughts regarding treatment of chronic 47. Lightowler JV, Wedzicha JA, Elliott MW, Ram FS. Non-inva-
obstructive pulmonary disease. Med Clin North Am 1996; sive positive pressure ventilation to treat respiratory failure
80:589-609. resulting from exacerbations of chronic obstructive pul-
34. Thompson WH, Nielson CP, Carvalho P, Charan NB, monary disease: Cochrane systematic review and meta-
Crowley JJ. Controlled trial of oral prednisone in outpa- analysis. Bmj 2003; 326:185.
tients with acute COPD exacerbation. Am J Respir Crit 48. Meyer TJ, Hill NS. Noninvasive positive pressure ventilation
Care Med 1996; 154:407-12. to treat respiratory failure. Ann Intern Med 1994; 120:760-70.
35. Davies L, Angus RM, Calverley PM. Oral corticosteroids in 49. Consensus conference report. Clinical indications for non-
patients admitted to hospital with exacerbations of chronic invasive positive pressure ventilation in chronic respiratory
obstructive pulmonary disease: a prospective randomised failure due to restrictive lung disease, COPD, and noctur-
controlled trial. Lancet 1999; 354:456-60. nal hypoventilation. Chest 1999; 116:521-34.
36. Niewoehner DE, Erbland ML, Deupree RH, Collins D, 50. Bott J, Carroll MP, Conway JH, Keilty SE, Ward EM, Brown
Gross NJ, Light RW, et al. Effect of systemic glucocorti- AM, et al. Randomised controlled trial of nasal ventilation
coids on exacerbations of chronic obstructive pulmonary in acute ventilatory failure due to chronic obstructive air-
disease. Department of Veterans Affairs Cooperative Study ways disease. Lancet 1993; 341:1555-7.
Group. N Engl J Med 1999; 340:1941-7. 51. Brochard L, Mancebo J, Wysocki M, Lofaso F, Conti G,
37. Maltais F, Ostinelli J, Bourbeau J, Tonnel AB, Jacquemet Rauss A, et al. Noninvasive ventilation for acute exacerba-
N, Haddon J, et al. Comparison of nebulized budesonide tions of chronic obstructive pulmonary disease. N Engl J
and oral prednisolone with placebo in the treatment of Med 1995; 333:817-22.
acute exacerbations of chronic obstructive pulmonary dis- 52. Kramer N, Meyer TJ, Meharg J, Cece RD, Hill NS.
ease: a randomized controlled trial. Am J Respir Crit Care Randomized, prospective trial of noninvasive positive pres-
Med 2002; 165:698-703. sure ventilation in acute respiratory failure. Am J Respir
38. Shepperd S, Harwood D, Jenkinson C, Gray A, Vessey M, Crit Care Med 1995; 151:1799-806.
Morgan P. Randomised controlled trial comparing hospital 53. Plant PK, Owen JL, Elliott MW. Early use of non-invasive
at home care with inpatient hospital care. I: three month ventilation for acute exacerbations of chronic obstructive
follow up of health outcomes. BMJ 1998; 316:1786-91. pulmonary disease on general respiratory wards: a multi-
39. Gravil JH, Al-Rawas OA, Cotton MM, Flanigan U, Irwin A, centre randomised controlled trial. Lancet 2000; 355:1931-5.
Stevenson RD. Home treatment of exacerbations of chron- 54. Rossi A, Gottfried SB, Higgs BD, Zocchi L, Grassino A,
ic obstructive pulmonary disease by an acute respiratory Milic-Emili J. Respiratory mechanics in mechanically venti-
assessment service. Lancet 1998; 351:1853-5. lated patients with respiratory failure. J Appl Physiol 1985;
40. Soderstrom L, Tousignant P, Kaufman T. The health and 58:1849-58.
cost effects of substituting home care for inpatient acute 55. Esteban A, Anzueto A, Alia I, Gordo F, Apezteguia C,
care: a review of the evidence. CMAJ 1999; 160:1151-5. Palizas F, et al. How is mechanical ventilation employed in
41. The COPD Guidelines Group of the Standards of Care the intensive care unit? An international utilization review.
Committee of the BTS. BTS guidelines for the manage- Am J Respir Crit Care Med 2000; 161:1450-8.
ment of chronic obstructive pulmonary disease. Thorax 56. American Thoracic Society. International Consensus
1997; 52 Suppl 5:S1-28. Conferences in Intensive Care Medicine: noninvasive posi-
42. Barbera JA, Reyes A, Roca J, Montserrat JM, Wagner PD, tive pressure ventilation in acute respiratory failure. Am J
Rodriguez-Roisin R. Effect of intravenously administered Respir Crit Care Med 2001; 163(1):283-91.
aminophylline on ventilation/perfusion inequality during 57. Plant PK, Owen JL, Elliott MW. Non-invasive ventilation in
recovery from exacerbations of chronic obstructive pul- acute exacerbations of chronic obstructive pulmonary dis-
monary disease. Am Rev Respir Dis 1992; 145:1328-33. ease: long term survival and predictors of in-hospital out-
43. Mahon JL, Laupacis A, Hodder RV, McKim DA, Paterson come. Thorax 2001; 56:708-12.
NA, Wood TE, et al. Theophylline for irreversible chronic 58. Keenan SP, Kernerman PD, Cook DJ, Martin CM,
airflow limitation: a randomized study comparing n of 1 tri- McCormack D, Sibbald WJ. Effect of noninvasive positive
als to standard practice. Chest 1999; 115:38-48. pressure ventilation on mortality in patients admitted with
44. Lloberes P, Ramis L, Montserrat JM, Serra J, Campistol J, acute respiratory failure: a meta-analysis. Crit Care Med
Picado C, et al. Effect of three different bronchodilators 1997; 25:1685-92.
during an exacerbation of chronic obstructive pulmonary 59. Purro A, Appendini L, De Gaetano A, Gudjonsdottir M,
disease. Eur Respir J 1988; 1:536-9. Donner CF, Rossi A. Physiologic determinants of ventilator
45. Murciano D, Aubier M, Lecocguic Y, Pariente R. Effects of dependence in long-term mechanically ventilated patients.
theophylline on diaphragmatic strength and fatigue in Am J Respir Crit Care Med 2000; 161:1115-23.
patients with chronic obstructive pulmonary disease. N 60. Torres A, Reyes A, Roca J, Wagner PD, Rodriguez-Roisin
Engl J Med 1984; 311:349-53. R. Ventilation-perfusion mismatching in chronic obstructive
46. Emerman CL, Connors AF, Lukens TW, May ME, Effron D. pulmonary disease during ventilator weaning. Am Rev
Theophylline concentrations in patients with acute exacer- Respir Dis 1989; 140:1246-50.
bation of COPD. Am J Emerg Med 1990; 8:289-92.

MANAGEMENT OF COPD 97
GOLD_WR_05 8/18/05 12:56 PM Page 98

61. Beydon L, Cinotti L, Rekik N, Radermacher P, Adnot S, 75. Man WD, Polkey MI, Donaldson N, Gray BJ, Moxham J.
Meignan M, et al. Changes in the distribution of ventilation Community pulmonary rehabilitation after hospitalisation
and perfusion associated with separation from mechanical for acute exacerbations of chronic obstructive pulmonary
ventilation in patients with obstructive pulmonary disease. disease: randomised controlled study. BMJ. 2004 Nov
Anesthesiology 1991; 75:730-8. 20;329(7476):1209.
62. Nava S, Ambrosino N, Clini E, Prato M, Orlando G, Vitacca 76. Saint S, Bent S, Vittinghoff E, Grady D. Antibiotics in chron-
M, et al. Noninvasive mechanical ventilation in the weaning ic obstructive pulmonary disease exacerbations. A meta-
of patients with respiratory failure due to chronic obstruc- analysis. JAMA 1995; 273(12):957-60.
tive pulmonary disease. A randomized, controlled trial. Ann 77. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES,
Intern Med 1998; 128:721-8. Harding GK, Nelson NA. Antibiotic therapy in exacerba-
63. Esteban A, Frutos F, Tobin MJ, Alia I, Solsona JF, Valverdu tions of chronic obstructive pulmonary disease. Ann Intern
I, et al. A comparison of four methods of weaning patients Med 1987; 106(2):196-204.
from mechanical ventilation. Spanish Lung Failure 78. Stockley RA, O'Brien C, Pye A, Hill SL. Relationship of
Collaborative Group. N Engl J Med 1995; 332:345-50. sputum color to nature and outpatient management of
64. Brochard L, Rauss A, Benito S, Conti G, Mancebo J, Rekik acute exacerbations of COPD. Chest 2000; 117(6):1638-45.
N, et al. Comparison of three methods of gradual withdraw- 79. Nouira S, Marghli S, Belghith M, Besbes L, Elatrous S,
al from ventilatory support during weaning from mechani- Abroug F. Once daily oral ofloxacin in chronic obstructive
cal ventilation. Am J Respir Crit Care Med 1994; 150:896-903. pulmonary disease exacerbation requiring mechanical ven-
65. Hilbert G, Gruson D, Portel L, Gbikpi-Benissan G, tilation: a randomised placebo-controlled trial. Lancet 2001;
Cardinaud JP. Noninvasive pressure support ventilation in 358(9298):2020-5.
COPD patients with postextubation hypercapnic respiratory 80. Pela R, Marchesani F, Agostinelli C, Staccioli D, Cecarini L,
insufficiency. Eur Respir J 1998; 11:1349-53. Bassotti C, Sanguinetti CM. Airways microbial flora in
66. Kessler R, Faller M, Fourgaut G, Mennecier B, COPD patients in stable clinical conditions and during
Weitzenblum E. Predictive factors of hospitalization for exacerbations: a bronchoscopic investigation. Monaldi Arch
acute exacerbation in a series of 64 patients with chronic Chest Dis 1998;53(3):262-7.
obstructive pulmonary disease. Am J Respir Crit Care Med 81. Soler N, Torres A, Ewig S, Gonzalez J, Celis R, El-Ebiary
1999; 159:158-64. M, Hernandez C, Rodriguez-Roisin R. Bronchial microbial
67. Mushlin AI, Black ER, Connolly CA, Buonaccorso KM, patterns in severe exacerbations of chronic obstructive pul-
Eberly SW. The necessary length of hospital stay for monary disease (COPD) requiring mechanical ventilation.
chronic pulmonary disease. JAMA 1991; 266:80-3. Am J Respir Crit Care Med 1998;157(5 Pt 1):1498-1505.
68. Cotton MM, Bucknall CE, Dagg KD, Johnson MK, 82. Fagon JY, Chastre J, Trouillet JL, Domart Y, Dombret MC,
MacGregor G, Stewart C, et al. Early discharge for patients Bornet M, Gibert C. Characterization of distal bronchial
with exacerbations of chronic obstructive pulmonary dis- microflora during acute exacerbation of chronic bronchitis.
ease: a randomized controlled trial. Thorax 2000; 55:902-6. Use of the protected specimen brush technique in 54
69. Hughes SL, Weaver FM, Giobbie-Hurder A, Manheim L, mechanically ventilated patients. Am Rev Respir Dis 1990;
Henderson W, Kubal JD, et al. Effectiveness of team-man- 142(5):1004-8.
aged home-based primary care: a randomized multicenter 83. Miravitlles M, Espinosa C, Fernandez-Laso E, Martos JA,
trial. Jama 2000; 284:2877-85. Maldonado JA, Gallego M. Relationship between bacterial
70. Hermiz O, Comino E, Marks G, Daffurn K, Wilson S, Harris flora in sputum and functional impairment in patients with
M. Randomised controlled trial of home based care of acute exacerbations of COPD. Study Group of Bacterial
patients with chronic obstructive pulmonary disease. Bmj Infection in COPD. Chest 1999; 116(1):40-6.
2002; 325:938. 84. Eller J, Ede A, Schaberg T, Niederman MS, Mauch H,
71. Stoller JK, Lange PA. Inpatient management of chronic Lode H. Infective exacerbations of chronic bronchitis: rela-
obstructive pulmonary disease. Respir Care Clin N Am tion between bacteriologic etiology and lung function.
1998; 4:425-38. Chest 1998; 113(6):1542-8.
85. Balter MS, La Forge J, Low DE, Mandell L, Grossman RF.
72. Peach H, Pathy MS. Follow-up study of disability among
Canadian guidelines for the management of acute exacer-
elderly patients discharged from hospital with exacerba-
bations of chronic bronchitis. Can Respir J 2003; 10 Suppl
tions of chronic bronchitis. Thorax 1981; 36:585-9.
B:3B-32B.
73. Aaron SD, Vandemheen KL, Hebert P, Dales R, Stiell IG,
86. Wilson R, Allegra L, Huchon G, Izquierdo JL, Jones P,
Ahuja J, Dickinson G, Brison R, Rowe BH, Dreyer J, Yetisir
Schaberg T, Sagnier PP. Short-term and long-term out-
E, Cass D, Wells G. Outpatient oral prednisone after
comes of moxifloxacin compared to standard antibiotic
emergency treatment of chronic obstructive pulmonary
treatment in acute exacerbations of chronic bronchitis.
disease. N Engl J Med. 2003 Jun 26;348(26):2618-25.
Chest 2004; 125(3):953-64.
74. Barr RG, Rowe BH, Camargo CA. Methylxanthines for
87. Martinez FJ, Grossman RF, Zadeikis N, Fisher AC, Walker
exacerbations of chronic obstructive pulmonary disease.
K, Ambruzs ME, Tennenberg AM. Patient stratification in
Cochrane Database Syst Rev. 2003;(2):CD002168.
the management of acute bacterial exacerbation of chronic
(Update of: Cochrane Database Syst Rev.
bronchitis: the role of levofloxacin 750 mg. Eur Respir J
2001;(1):CD002168.)
2005; 25(6):1001-10

98 FUTURE RESEARCH
GOLD_WR_05 8/18/05 12:56 PM Page 99

CHAPTER

FUTURE

RESEARCH
GOLD_WR_05 8/18/05 12:56 PM Page 100

CHAPTER 6: FUTURE RESEARCH


A better understanding of the molecular and cellular ❥ Longitudinal studies demonstrating the course of
pathogenic mechanisms of COPD should lead to many COPD are needed in a variety of populations exposed to
new directions for both basic and clinical investigations. various risk factors. Such studies would provide insight
Improved methods of early detection, new approaches for into the pathogenesis of COPD, identify additional genetic
interventions through targeted pharmacotherapy, possible bases for COPD, and identify how genetic risk factors
means to identify the “susceptible” smoker, and more interact with environmental risk factors in specific patient
effective means of managing exacerbations are needed. populations. Factors that determine why some, but not
all, smokers develop COPD need to be identified.
Some research recommendations and future program
goals are provided to stimulate the efforts of investigators ❥ Data are needed on the use, cost, and relative distribution
around the world. There are many additional avenues to of medical and non-medical resources for COPD, especially
explore. in countries where smoking and other risk factors are
prevalent. These data are likely to have some impact
❥ Until there is a better understanding of the causal on health policy and resource allocation decisions. As
mechanisms of COPD, an absolutely rigid definition of options for treating COPD grow, more research will be
COPD, and its relationship to other obstructive airways needed to help guide health care workers and health
diseases, will remain controversial. The defining budget managers regarding the most efficient and effective
characteristics of COPD should be better identified. ways of managing this disease. Methods and strategies
for implementation of COPD management programs in
❥ The stages and natural history of COPD vary from one developing countries will require special attention.
patient to another. The clinical utility of the four-stage
classification of severity used in the GOLD Report needs ❥ While spirometry is recommended to assess and
to be evaluated. monitor COPD, other measures need to be developed
and evaluated in clinical practice. Reproducible and
❥ Surrogate markers of inflammation, possibly derived inexpensive exercise-testing methodologies (e.g., stair-
from the analysis of sputum (cells, mediators, enzymes) climbing tests) suitable for use in developing countries
or exhaled condensates (lipid mediators, reactive oxygen need to be evaluated and their use encouraged.
species, cytokines), that may predict the clinical usefulness Spirometers need to be developed that can ensure e
of new management and prevention strategies for COPD conomical and accurate performance when a relatively
need to be developed. untrained operator administers the test.

❥ Information is needed about the cellular and molecular ❥ Since COPD is not fully reversible (with current
mechanisms involved in inflammation in stable COPD therapies) and slowly progressive, it will become ever
and exacerbations. Inflammatory responses in nonsmokers, more important to identify early cases as more effective
ex-smokers, and smokers with and without COPD should therapies emerge. Consensus on standard methods for
be compared. The mechanisms responsible for the detection and definition of early disease need to be
persistence of the inflammatory response in COPD should developed. Data to show whether or not screening is
be investigated. Why inflammation in COPD is poorly effective in directing management decisions in COPD
responsive to glucocorticosteroids and what treatments outcomes are required.
other than glucocorticosteroids are effective in suppressing
inflammation in COPD are research topics that could lead ❥ Primary prevention of COPD is one of the major
to new treatment modalities. objectives of GOLD. Investigations into the most cost-
effective ways to reduce the prevalence of tobacco smoking
❥ Standardized methods for tracking trends in COPD in the general population and more specifically in young
prevalence, morbidity, and mortality over time need to be people are very much needed. Strategies to prevent people
developed so that countries can plan for future increases from starting to smoke and methods for smoking cessation
in the need for health care services in view of predicted require constant evaluation and improvement. Research
increases in COPD. This need is especially urgent in is required to gauge the impact and reduce the risk from
developing countries with limited health care resources. increasing air pollution, urbanization, recurrent childhood

100 FUTURE RESEARCH


GOLD_WR_05 8/18/05 12:56 PM Page 101

infections, occupational exposures, and use of local Mediator antagonists: Attention has largely focused
cigarette equivalents. Programs designed to reduce on mediators involved in recruitment and activation of
exposure to biomass fuel in countries where this is used neutrophils, and reactive oxygen species. In this category
for cooking and domestic heating should be explored in an are the LTB4 antagonists, lipoxygenase inhibitors,
effort to reduce exposure and improve ventilation in homes. chemokine inhibitors, and TNF- inhibitors.

❥ The specific components of effective education for Antioxidants: Oxidative stress is increased in patients
COPD patients need to be determined. It is not known, with COPD, particularly during exacerbations. Oxidants
for example, whether COPD patients should be given an are present in cigarette smoke and are produced
individual management plan, or whether these plans are endogenously by activated inflammatory cells, including
effective in reducing health care costs or improving the neutrophils and alveolar macrophages, suggesting that
outcomes of exacerbations. Developing and evaluating antioxidants may be of use in therapy for COPD.
effective tools for physician education concerning prevention,
diagnosis, and management of COPD will be important in Anti-inflammatory drugs: The limited value of glucocor-
view of the increasing public health problem presented by ticosteroids in reducing inflammation in COPD suggests
COPD. that novel types of nonsteroidal anti-inflammatory treatment
may be needed. There are several new approaches to
❥ Studies are needed to determine whether education is anti-inflammatory treatment in COPD including, for example,
an essential component of pulmonary rehabilitation. The phosphodiesterase inhibitors, transcription factor NF-B
cost effectiveness of rehabilitation programs has not been inhibitors, and adhesion molecule blockers.
assessed and there is a need to assess the feasibility,
resource utilization, and health outcomes of rehabilitation Proteinase inhibitors: There is compelling evidence
programs that are delivered outside the major teaching that an imbalance between proteinases that digest elastin
hospital setting. Criteria for selecting individuals for (and other structural proteins) and antiproteinases that
rehabilitation should be evaluated, along with methods to protect against this digestion exists in COPD.
modify programs to suit the needs of individual patients. Considerable progress has been made in identifying the
enzymes involved in elastolytic activity in emphysema
❥ Collecting and evaluating data to classify COPD and in characterizing the endogenous antiproteinases
exacerbations by severity would stimulate standardization that counteract this activity, including neutrophil elastase
of this outcome measure that is so frequently used in inhibitors, cathepsin G and proteinase 3 inhibitors, and
clinical trials. Further exploration of the ethical principles matrix metalloproteinase inhibitors. Other serine pro-
of life support and greater insight into the behavioral teinase inhibitors (serpins), such as elafin, may also be
influences that inhibit discussion of such intangible issues important in counteracting elastolytic activity in the lung.
are needed, along with studies to define the needs of
end-stage COPD patients. Mucoregulators: It may be important to develop drugs
that inhibit the hypersecretion of mucus, without
❥ There is a pressing need to develop drugs that control suppressing the normal secretion of mucus or impairing
symptoms and prevent the progression of COPD. Some mucociliary clearance. There are several types of
progress has been made and there are several classes of mucoregulatory drugs in development including tachykinin
drugs that are now in preclinical and clinical development antagonists, sensory neuropeptide inhibitors, mediator
for use in COPD patients. and enzyme inhibitors, mucin gene suppressors, mucolytic
agents, macrolide antibiotics, and purinoceptor blockers.
Bronchodilators: Bronchodilators are the mainstay of
symptomatic therapy and new short-acting and long-acting Alveolar repair: A major mechanism of airway obstruction
bronchodilators are anticipated. With the recognition that in COPD is loss of elastic recoil due to proteolytic
there are different subtypes of muscarinic receptors, destruction of the lung parenchyma. Thus, it seems
there has been a search for more selective antagonists. unlikely that this obstruction can be reversed by drug
Tiotropium bromide, a new drug in advanced clinical trials, therapy, although it might be possible to reduce the rate
is a quaternary ammonium compound like ipratropium of progression by preventing the inflammatory and
bromide, but with the unique property of kinetic selectivity enzymatic disease processes. It is even possible that
and very long duration of action. Selective phosphodi- drugs might be developed to stimulate regrowth of alveoli.
esterase type IV inhibitors might combine bronchodilator Retinoic acid increases the number of alveoli in rats and,
and anti-inflammatory activity. remarkably, reverses the histological and physiological

FUTURE RESEARCH 101


GOLD_WR_05 8/18/05 12:56 PM Page 102

changes induced by elastase treatment. The molecular


mechanisms involved and whether this can be extrapolated
to humans are not yet known. Several retinoic acid
receptor subtype agonists have now been developed that
may have a greater selectivity for this effect. Hepatocyte
growth factor (HGF) has a major effect on the growth of
alveoli in the fetal lung, and it is possible that in the future
drugs might be developed that switch on responsiveness
to HGF in adult lung or mimic the action of HGF.

Route of delivery: Many inhalers that deliver bron-


chodilators have been optimized to deliver drugs to the
respiratory tract in asthma. Methods to quickly and safely
deliver medications to target sites of inflammation and
tissue destruction in COPD need to be evaluated.

102 FUTURE RESEARCH


GOLD_WR_05 8/18/05 12:56 PM Page 103

Note: This segment on Outcomes and Markers in COPD is presented by the GOLD Science
Committee as an Abstract to the GOLD Workshop Report (updated 2005) as a “work in
progress.” Comments may be sent to the GOLD Science Committee (shurd@prodigy.net).
The GOLD Science Committee intends to include segments from this document in the full
revision of the report, scheduled to appear in mid-2006.

OUTCOMES AND MARKERS IN COPD


TABLE OF CONTENTS
PARTICIPANTS ................................................................................................................................................................. 2

PREFACE.......................................................................................................................................................................... 3

I. INTRODUCTION......................................................................................................................................................... 4

II. TERMINOLOGY AND DEFINITIONS .......................................................................................................................... 4

A. Clinical outcome.................................................................................................................................................. 4
B Marker................................................................................................................................................................. 4
C. Relationship between markers and outcomes.................................................................................................... 5
D. Clinical outcomes, markers and modifiers in COPD........................................................................................... 6
E Worked example ................................................................................................................................................. 6

III. CLINICAL OUTCOMES ............................................................................................................................................... 6

A. Mortality ............................................................................................................................................................. 6
B. Symptoms and quality of life .............................................................................................................................. 7
C. Exercise tolerance ............................................................................................................................................. 7
D. Exacerbations and acute respiratory failure ...................................................................................................... 7
E. Weight loss......................................................................................................................................................... 7
F. Use of health care and non-health care resources ............................................................................................ 8

IV. MARKERS.................................................................................................................................................................... 8

A. Mortality .............................................................................................................................................................. 8
B. Symptoms and health status............................................................................................................................... 8
C. Exacerbations and acute respiratory failure ....................................................................................................... 9
D. Lung function....................................................................................................................................................... 9
E. Exercise capacity ................................................................................................................................................ 10
F. Weight Loss ......................................................................................................................................................... 10
G. Imaging ............................................................................................................................................................... 10
H. Resource utilization............................................................................................................................................. 11
I. Biomarkers .......................................................................................................................................................... 11
J. Composite markers.............................................................................................................................................. 11

V. SUMMARY AND CONCLUSIONS ............................................................................................................................... 11

TABLE 1. PROPERTIES OF THE IDEAL MARKER ......................................................................................................... 12


TABLE 2. OUTCOME MEASURES FOR COPD............................................................................................................... 13
TABLE 3. MARKERS FOR STABLE COPD ...................................................................................................................... 14
TABLE 4: MARKERS FOR EXACERBATIONS OF COPD .............................................................................................. 15

REFERENCES.................................................................................................................................................................. 15

OUTCOME AND MARKERS 103


GOLD_WR_05 8/18/05 5:19 PM Page 104

OUTCOMES AND MARKERS IN COPD


PARTICIPANTS
EXECUTIVE COMMITTEE: L. Fabbri, Chair: A. Agusti, S. Buist, P. Calverley, S. Hurd, C. Jenkins, P. Jones, K. Rabe,
R. Rodriguez-Roisin

WORKING GROUPS: Provided materials to develop the report and invited to participate
in review.

A. Mortality: B. Celli, Chair, Boston, Massachusetts, US


J. Vestbo, Hvidore, Denmark
E. Wouters, Maastricht, The Netherlands
T. Oga, Kyoto, Japan

B. Exacerbation, respiratory failure: C. Jenkins, Chair, NSW Australia


R. Rodriguez Roisin, Barcelona, Spain
S. Sethi, Buffalo, New York, US
J. A. Wedzicha, London, UK
A. Rossi, Verona, Italy

C. Symptoms, quality of life: S. Rennard, Chair, Omaha, Nebraska, US


H. Schunemann, Buffalo, New York, US
P. Jones, Tonbridge, Kent, UK

D. Lung Function: L. Fabbri, Chair, Modena, Italy


A. S. Buist, Portland, Oregon, US
P. Sterk, Leiden, The Netherlands

E. Exercise: P. Calverley, Chair, Liverpool, UK


K. Nishimura, Kyoto, Japan
Jose Albert Jardim, Sao Paulo, Brazil

F. Imaging: K. Rabe, Chair, Leiden, The Netherlands

G. Biomarkers: S. Rennard, Chair, Omaha, Nebraska, US


A. G. Agusti, Palma de Mallorca, Spain

H. Health Care Utilization: S. Sullivan, Seattle, Washington, US

GOLD NATIONAL LEADERS: Invited to participate in review.

104 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 105

OUTCOMES AND MARKERS IN COPD


PREFACE recommendations are expected to be incorporated into the
management section of the revised GOLD guidelines that
The lack of generally accepted outcome measures in will be available by the end of 2006.
COPD (other than FEV1) that can be used as criteria
for the evaluation of novel treatments and for approval
of new medications greatly hinders research and clinical
practice. This problem has been long recognized by Leonardo Fabbri, MD
research scientists, clinical investigators, industry Chair, GOLD Executive Committee
representatives, and regulatory authorities worldwide. July 31, 2005

In 2001, the Global Initiative for Chronic Obstructive Lung


Disease (GOLD) developed, and widely distributed,
evidence-based guidelines for COPD therapy titled Global
Strategy for Diagnosis, Management and Prevention of
COPD1. These guidelines have been updated each year
2 to assure that recommendations for COPD treatment
and management are based on current published literature.
In 2004, under the leadership of Professor Romain
Pauwels, GOLD convened an expert panel to review data
on outcome measures and to identify those that could be
used to evaluate management of COPD as described in
the GOLD guidelines. The panel was also asked to
provide guidance about additional research that may be
needed to confirm the validity of the use of other
outcome measures.

With the GOLD guidelines as the foundation of this


project, and the consultation and support of respected
experts from several regions of the world, we hope that
the recommendations provided in this report will stimulate
discussion in the scientific community as well as additional
research to fill the several gaps in knowledge. We strongly
believe that the process initiated by this “working” document
will eventually benefit clinical practice, clinical research
concerning new COPD medications, regulatory decision
making, and patient welfare.

We are indebted to Dr. Romain Pauwels for initiation of


this project. His untimely death occurred before he could
have significant input into this report. We are grateful for
the expert consultation provided in particular by Dr. Paul
Jones and Dr. Alvar Agusti who, along with other
colleagues on the panel, provided the recommendations
to the GOLD Executive Committee. During the 12-month
period beginning July 1, 2005, this report will be included
as an Appendix to the 2005 update of the Global
Strategy for Diagnosis, Management and Prevention of
COPD and posted on the GOLD website:
http://www.goldcopd.org for comments. The final

OUTCOME AND MARKERS 105


GOLD_WR_05 8/18/05 12:56 PM Page 106

I. INTRODUCTION B. Marker

Patients with COPD are heterogeneous in terms of their A marker is a measurement associated with one or
clinical presentation, co-morbidities, underlying lung more clinical outcomes.
pathology, disease severity, and rate of disease progression.
Thus it is highly unlikely that a single measure can
accurately assess the severity of COPD, predict patient The term “marker” is used in a number of contexts:
prognosis, and evaluate the effectiveness of therapy,
thereby measuring all the dimensions of the disease3. • Diagnostic marker - used as a dichotomous variable
Yet traditionally, the forced expiratory volume in one - present or absent. It may not be measured on a
second (FEV1) has been used extensively as a global dichotomous scale, but is assigned to one of two
measurement for COPD. While the long term excessive states, based on ranges defined from experience;
decline of FEV1 is the pathognomonic abnormality of e.g., alpha1-antitrypsin level, or FEV1 when used for
COPD and may indeed relate to mortality, FEV1 varies COPD diagnosis.
little over short periods of time, even during exacerbations,
and it relates weakly to other clinical manifestations such • Marker of disease severity - describes different
as quality of life or exercise tolerance. It is clear, therefore, levels of disease severity by categorizing levels of
that other measures (besides FEV1) need to be identified the marker into predefined ranges, e.g., Body Mass
and validated to allow a more complete and clinically Index (BMI), or FEV1 as used in GOLD staging.
relevant assessment of patients with COPD3.
• Marker of disease progression - used to assess
The terms “clinical outcome” and “marker” have been the course of the disease (e.g., rate of decline in
increasingly used over the past few years in relation to FEV1 or rate of deterioration in health status).
COPD, yet considerable confusion and misuse of these
terms exists. The goals of this document are to: • Marker of treatment effect - used to measure
response to treatment (e.g., dyspnea score, lean
• clarify the meaning of the terms “clinical outcome” and body mass, exercise capacity, health status, FEV1,
“marker” and suggest how these terms can contribute etc.). In clinical trials these markers are usually
to COPD management described in GOLD1; termed ‘outcome variables’.
• review the available evidence supporting the use of
different clinical outcomes/markers in the overall • Biomarker – a measurement of chemical or biological
assessment of a treatment or intervention in COPD material that reflects a disease process, e.g. a bio-
patients; marker for inflammation.
• identify existing gaps of knowledge where further
research is required. • Surrogate marker – applies when one marker is
used as a substitute for the marker of primary interest,
II. TERMINOLOGY AND DEFINITIONS e.g., High resolution computed tomography (HRCT)
scan densitometry measurement as a surrogate
A. Clinical Outcome marker for the presence of emphysema.

Regardless of these different uses, the ideal marker


A clinical outcome is a consequence of COPD
should have the properties shown in Table 1.
experienced by the patient (symptoms, weight loss,
exorcise intolerance, exacerbations, health care
C. Relationship between markers and outcomes
resource use, mortality).
The relationship between markers and outcomes is not
The meaning of the term “outcome” varies depending straightforward. The following factors need to be
on the context. For instance, in clinical trials, the term considered:
“outcome variable” refers to the main variable of interest,
irrespective of its nature or type (e.g., FEV1, FEV1 • A clinical outcome may have multiple markers (e.g.,
decline, exacerbations). In this document, “outcome” will BMI, FEV1 and exercise capacity are all independent
be used in the context of the clinical assessment of predictors of mortality).
COPD.

106 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 107

• The relationship between a marker and a clinical • Surrogate marker for exercise capacity: FEV1
outcome may be altered by modifiers that are not • Modifier: co-morbidity (e.g., heart failure)
directly related to the disease but may have a
significant impact on its clinical outcomes. Modifiers The correlation between exercise capacity and FEV1 is
in COPD include co-morbidity, level of social support, not always strong. This is often the case with surrogate
and ease of access to the health care system. markers, and, as a general rule, they should be used only
when it is not possible to use the relevant marker. In a
• A small number of markers may be so well clinical trial, exercise capacity may be used as the primary
characterized and understood that they can substitute outcome variable, because it is a valid marker of exercise
effectively for a clinical outcome and become an tolerance (the clinical outcome of interest in this example).
outcome of treatment in itself. For instance, in the
treatment of cardiovascular disease a reduction in III. CLINICAL OUTCOMES
blood pressure (a marker) has become an accepted
clinical outcome since it is known to reduce the A. Mortality
probability of cardiovascular morbidity and mortality.
Mortality as a clinical outcome for COPD can be measured,
• Occasionally a marker may be used both as a marker is meaningful, and is obviously clinically relevant to the
of disease severity and as a clinically relevant disease process, in particular to the end-stage disease7.
outcome in itself. One example, weight loss, is A drawback to using mortality as an outcome in COPD is
particularly prevalent among patients with severe that it is often not listed on the death certificate, or may
COP4, but influences prognosis independently of the only be listed as a contributory cause of death. However,
level of lung function impairment5,6. Thus, it is both in clinical studies, particularly those in which participants
an outcome that affects the patient (e.g., in terms are followed over time and in which there is supporting
of body image) and a marker of underlying disease clinical information about markers of disease severity,
activity. mortality provides a very important clinical outcome
measure. In studies that use COPD mortality as a clinical
D. Clinical outcomes, markers and modifiers in COPD outcome, an adequate adjudication process for all deaths
must be followed so that COPD (as either a primary or a
Tables 2-4 summarize currently accepted clinical outcomes, contributory cause of death) is coded as accurately as
markers, and modifiers in COPD, and the potential use of possible8.
different markers in patients with stable and exacerbated
COPD. Once appropriately validated, some current B. Symptoms and quality of life
markers (e.g., HRCT) may become clinical outcomes in
themselves (as in the example of blood pressure cited The most frequent symptoms in COPD patients are
above). dyspnea, cough, sputum production, and fatigue.
Although fatigue is poorly specific for COPD, it has a
While clinical trials in patients with COPD have mainly very high prevalence but is rarely reported spontaneously
focused on changes in lung function as a marker of by COPD patients9. Symptoms contribute significantly
treatment effect and/or disease progression, the to other relevant clinical outcomes such as disability,
importance of measuring clinical outcomes such as restriction of normal daily activities, and emotional and
symptoms, exacerbations, and health-related quality of social disturbances. In turn, symptoms impair quality of
life (and their associated markers) is gaining increasing life, although the impact will be unique to each patient.
recognition because these outcomes are important to
patients. Lung function is only weakly related to these C. Exercise tolerance
outcomes (i.e., it is a poor surrogate marker).
The ability to exercise is significantly impaired in many
E. Worked example COPD patients and is an important determinant of
health-related quality of life10,11. as it impairs the ability to
A simple worked example of clinical outcomes, markers, carry out daily activities. The ability of exercise to provoke
surrogate markers, and modifiers: breathlessness is used in the Medical Research Council
(MRC) dyspnea scale to estimate symptom intensity12.
• Clinical outcome: exercise tolerance It is difficult to make reliable measurements of a patient’s
• Marker: exercise capacity measured in laboratory daily activity, so physiological measurements in the

OUTCOME AND MARKERS 107


GOLD_WR_05 8/18/05 12:56 PM Page 108

exercise laboratory are used as markers of impaired Emergency treatment data can be obtained from the
activity. Exercise capacity is not only a marker of exercise patient or care-giver, and from clinical or billing records25.
tolerance, but also an important predictor of mortality13,14. Data on preventive pharmacotherapy, diagnostic
investigations, and clinical follow-up are required to
D. Exacerbations and acute respiratory failure supplement data on emergency treatments in order to
provide a more comprehensive assessment of health
A COPD exacerbation is a sustained worsening of the resource use and costs. Assessment of patient and
patient’s condition from the stable state and beyond normal care-giver travel and waiting time, disability, absence from
day-to-day variation that is acute in onset and may warrant work, and productivity while at work comprise additional
additional treatment15. Patients with exacerbations of and important non-health care resource consumption
COPD typically present with increased breathlessness measures in COPD20.
with or without cough, changes in sputum volume and
purulence, wheezing, and chest tightness. Exacerbations IV. MARKERS
are an important clinical outcome of the disease as well
as an important marker of disease severity. A. Mortality

Severe exacerbations may be accompanied by acute To establish a precise cause of death is difficult for chronic
respiratory failure, defined as decreased arterial PO2 diseases (including COPD) that often are associated with
(arterial deoxygenation) with or without increased arterial other diseases. While all-cause mortality in COPD can
PCO2. Acute respiratory failure is a common clinical be assessed, death caused specifically by COPD heavily
outcome in severe exacerbations of COPD. Acute relies on accuracy of death certificate. Predictors of
respiratory failure is perceived by the patient as severe mortality from COPD include lung function (FEV1, forced
dyspnea often associated with agitation, confusion, vital capacity [FVC], inspiratory capacity/total lung capacity
tachycardia and sweating15. Mortality ranges between [IC/TLC]), blood gases (both PaO2 and PaCO2), respiratory
11%16 and 20%17 in patients needing mechanical ventilation. symptoms26, exercise capacity, BMI6, exacerbations and
combinations27,28. Of these, lung function is a marker of
E. Weight loss all-cause mortality and is associated with COPD mortality
even in the early stages of disease7.
Patients with moderate to severe COPD have a depletion
of fat-free mass, particularly skeletal muscle, that is B. Symptoms and health status
reflected by weight loss18,19. Weight loss is a predictor
of mortality in patients with COPD, and survival may Dyspnea is currently the only COPD symptom that can
improve with an increase in body-mass index. In addition be measured in a standardized manner, through the use
to the relation of low body weight and mortality, weight of Borg or Visual Analogue Scales usually applied during
loss is an important determinant of impaired muscle laboratory exercise tests. The Borg scale has standardized
strength, exercise capacity, exercise response, and health descriptors that allow more direct comparisons between
status, as well as increased morbidity (e.g., recurrent studies than Visual Analogue Scale scores. Other
exacerbations and readmission to hospital) in patients measures of dyspnea do not assess this symptom directly,
with COPD18,19. as they quantify self-reported activity limitation in daily
life. All dyspnea instruments listed in Table 3 (except the
F. Use of health care and non-health care resources Transitional dyspnea index [TDI]) can distinguish between
different degrees of breathlessness-induced disability,
Between 60 and 75 percent of medical expenditures for although the MRC scale is simplest and most widely
COPD are a direct consequence of exacerbations20-23, so used. The TDI and University of California San Diego
use of health care resources is an important outcome in (UCSD) instruments can respond to changes with
COPD representing treatment failure and progression of treatment but there are currently too few data to make
disease24. In clinical trials, use of emergency treatment, an assessment as to whether they can track long-term
alone or in combination with symptom and lung function disease progression29.
data, is customarily used to characterize an exacerbation
especially when the primary study outcome is reduction Health-related quality of life is a clinical outcome of
in the frequency or time to an exacerbation event. COPD that will be unique to each patient, so it cannot
be quantified in a standardized manner. Instead, health
status questionnaires are used as markers of the impact

108 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 109

of the disease on patients’ health, daily life and sense of D. Lung function
well-being. All three health status questionnaires (chronic
respiratory questionnaire [CRQ], St. Georges respiratory Lung function may act as a marker for clinical outcomes
questionnaire [SGRQ], 36 item short form [SF-36]) can such as symptoms, quality of life, exercise tolerance,
distinguish between different degrees of severity. The health care utilization, and mortality1,38. However, lung
disease-specific CRQ and SGRQ are sensitive to function measures relate weakly to these clinical
treatment, but the generic SF-36 is not consistently outcomes30. Lung function measures have been used
responsive to worthwhile therapeutic effects. The SGRQ extensively for the diagnosis and assessment of severity
and SF-36 have been shown to be responsive to long- of COPD1; FEV1 is the most readily available and
term disease progression, but similar evidence is not reproducible. Other parameters of lung function add
currently available for the CRQ30. little to FEV1, being either less reproducible or less
sensitive39,40. Post-bronchodilator FEV1 and FEV1/FVC
C Exacerbations and acute respiratory failure are essential markers for the diagnosis and assessment
of severity of COPD1. Because of its unimodal distribution
Exacerbations of COPD are clinical outcomes when and poor reproducibility, short-term reversibility testing
evaluating the impact of interventions. Exacerbation to bronchodilator or glucocorticosteroids add little to
frequency and severity are also used as markers of baseline lung function for diagnosis41,42, prediction of
COPD severity, progression, impact on quality of life, disease progression15, or response to treatment43.
and mortality16,31. Exacerbations are more frequent and
severe in advanced disease32, and exacerbation frequency Repeated measurements of FEV1 over a period of time
is related to the decline in quality of life experienced have been used to study the natural progression of
by the COPD patient30,33,34. There are a few clinical disease44,45. Follow-up studies have shown that the annual
classifications of exacerbation severity, mostly ranging decline in post-bronchodilator FEV1 may be more
from mild to life-threatening, and based essentially on reproducible than pre-bronchodilator as a parameter of
either symptom-driven35 or event-driven definitions36. lung function to assess progression45,46. Additional lung
function measures such as lung volumes and capacities
Major difficulties are currently encountered in studies (residual volume [RV], functional residual capacity [FRC],
that use exacerbations as endpoint primary outcome for inspiratory capacity [IC], and total lung capacity [TLC]),
assessing interventions because of the lack of a widely carbon monoxide diffusion capacity, and arterial blood
acknowledged definition of COPD exacerbations. In most gases may be helpful to assess severity (e.g., severe
studies, exacerbations have been defined on the basis COPD, exacerbations), and to predict the response to
of increase in symptoms, requiring patient perception specific treatments (e.g., lung volume reduction
and a reaction to this perception. Both vary significantly surgery)19,47, but have not been shown to add much to
between patients and may be influenced by a number of FEV1, being either less reproducible or less sensitive
modifiers, such as access to the health care system, and
the presence or absence of family or social support. E. Exercise capacity

Definitions of exacerbations based on the need for therapy Exercise capacity is a marker for clinical outcomes such
can be useful indicators of severity, but may also be as symptoms, quality of life, exercise tolerance, health
insensitive in some settings given that exacerbation care utilization, and mortality. Exercise capacity can be
therapies vary throughout the world. Patients with COPD evaluated by making detailed physiological measurements
exacerbations do not always seek medical care and are in the exercise laboratory (minute ventilation, breathing
increasingly using self-management strategies that may pattern, oxygen consumption, carbon dioxide production,
exclude visiting the primary care physician unless critically oxygen saturation, and oxygen pulse - all during exercise)
ill. When a hospital admission occurs, objective measures or by using simpler field tests where the duration of
of severity may be included. Symptoms, volume and exercise or the distance covered in a fixed time period is
color of sputum, use of health resources, lung function, recorded (e.g., six-minute walking test). Measures of
and blood gases may be considered the most relevant exercise capacity are close to the ideal marker (Table 1),
markers of COPD exacerbations (Table 4). Arterial pH as they have good validity, specificity, reliability,
and blood gases are the only proven markers of acute repeatability48, predictive ability14, discriminative ability
respiratory failure in COPD37. Oxygen saturation by pulse and evaluative ability47.
oximetry can also be used, although less accurately, and
does not provide information on arterial PCO2.

OUTCOME AND MARKERS 109


GOLD_WR_05 8/18/05 12:56 PM Page 110

F. Weight loss I. Biomarkers

Weight loss is a marker for clinical outcomes such as COPD is characterized by chronic inflammation56.
Inflammatory cells (e.g., T cells, neutrophils, and
symptoms, quality of life, exercise tolerance, health care
eosinophils), mediators (e.g., IL-8, TNFa, LTB4, proteases
utilization, and mortality. Serial measurements of body
and antiproteases, C-reactive protein [CRP]), and
weight can disclose progressive involuntary weight loss,
components of exhaled air or condensate involved in this
generally considered to be clinically relevant when it complex inflammatory cascade have been identified as
exceeds 5% over a month or 10% over 6 months49. potential biomarkers of the disease process, and have
Actual body weight can be related to the ideal body- been examined as markers for diagnosis57, assessment of
weight as derived from height, frame size, and gender. severity of stable COPD58, diagnosis/assessment of COPD
Nutritional depletion is generally and arbitrarily defined exacerbations59, and evaluation of the effect of treatment60,61.
as body weight of less than 90% of the ideal4. Body However, no single or combination of biomarkers has yet
weight can be corrected for body size by calculation of been identified that can be reliably used in clinical practice
body-mass index; a value lower than 20 kg/m2 is generally in the diagnosis, staging, or monitoring of COPD.
taken as abnormal4,50. The assessment of nutritional
status according to body weight provides no qualitative J. Composite markers
information on body tissues. A fat-free-mass index
(fat-free mass in kg divided by height squared) of less Because COPD is a multi-system, multi-component
than 16 kg/m2 in men and 15 kg/m2 in women is taken disease62, there has been increased interest in the use
as an indicator of active body-tissue depletion18,19. of composite markers that reflect the overall effect of the
disease. Two markers in particular may fulfill this function:
G. Imaging exercise testing, which reflects cardiopulmonary and
skeletal muscle function, and health status measurements,
Chest imaging can provide useful information for diagnosis which assess a wide range of symptomatic effects of the
and disease severity51. The chest x-ray is seldom disease. A composite score derived from four established
diagnostic in COPD, but is valuable in the differential clinical markers - BMI, MRC Dyspnea Grade, FEV1 and
6-minute walking distance (all included in Table 3) - has
diagnosis to exclude other diseases. A chest x-ray
been created and validated (the BODE Index: Body-
may also be helpful in phenotyping COPD in term of
mass index (B), the degree of airflow obstruction (O),
bronchiolitis or emphysema52. There is no evidence of the
dyspnea (D), exercise capacity (E)27). The components
value of chest x-ray to assess disease progression or of this instrument are all markers of mortality, and this
treatment effect. HRCT scan is progressively replacing instrument validated against mortality proved to be a
the regular chest x-ray for differential diagnosis of COPD, better predictor than FEV1 alone.
phenotyping of COPD (bronchiolitis vs. emphysema), and
assessment of COPD severity. Densitometric analysis of V. SUMMARY AND CONCLUSIONS
a CT-scan or HRCT scan be used to assess the presence
and severity of emphysema53, and thus is potentially useful Based on available evidence, definitions of clinical outcome
in assessing the progression of the disease, and the effect and markers in COPD have been proposed. Most published
of specific treatments (e.g., retinoids) on progression of investigations have concentrated on COPD as a respiratory
emphysema54,55. disease, and particularly on respiratory symptoms and
lung function, although more comprehensive approaches
H. Resource utilization addressing health-related quality of life and exercise testing
have recently appeared. The judicious use of many of the
The presence and frequency of health care resource outcomes and markers described in this report should
utilization are good markers of COPD exacerbations, greatly enhance the development of new therapeutic
disease severity, and progression of disease22. In general, strategies that may eventually contribute to improve the
and for patients with chronic disease specifically, increasing management of COPD. The increasing evidence that
age and female gender are positively related to resource COPD is a multi-component, complex disease suggests the
consumption and costs. The general health status question need for the identification and use of more comprehensive
on the SF-36, when adjusted for age and gender, has clinical outcomes and more accurate markers or biomarkers
to assess disease severity, prognosis, and response to
been shown to predict mortality and health care resource
therapy. This should be an important goal for future
utilization.
clinical research investigations.

110 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 111

TABLE 1. PROPERTIES OF THE IDEAL MARKER

PROPERTY DEFINITION
Validity Strong relationship to underlying disease mechanisms and well-being

Specificity Absence of confounding effects of co-morbidities or other factors

Reliability Performs consistently in different settings.

Repeatability Measurements do not change in the stable state

Predictive ability Predicts clinical outcomes

Discriminative ability Identifies differences in severity between patients

Evaluative ability Sensitive to changes within patients.

Simplicity Routine or research procedure

Cost-effective Savings from improved management offset the cost

TABLE 2. OUTCOME MEASURE FOR COPD


Outcome Clinical Relevance Markers Modifiers

Mortality End of life FEV1 Co-morbidity


BMI Age
MRC dyspnea Social/family support
Exercise capacity Access to health care system
BODE
PaO2, PaCO2
Exacerbations
Health status
Symptoms Health status Health status Co-morbidity
Quality of Life Dyspnea Dyspnea scales

Exacerbations Mortality Previous frequency Co-morbidity


Health status FEV1 Age
Lung function decline PaCO2 Social/family support
Weight loss Bronchial colonization Access to health care system
Inflammatory markers
Exercise Tolerance Health status FEV1 Co-morbidity
Exacerbations PaO2 Age
BMI

Weight Loss Survival Weight Co-morbidity


Exercise tolerance PaO2,PaCO2 Age
Health status DLCO Social/family support
Exacerbations CT-emphysema

Health Care Utilization Health status FEV1 Age


Economic cost PaO2,PaCO2 Co-morbidity
BMI Active smoking
Exercise tolerance Social/family support
Health status Access to health care system

OUTCOME AND MARKERS 111


GOLD_WR_05 8/18/05 12:56 PM Page 112

112 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 113

TABLE 4. MARKERS FOR EXACERBATIONS OF COPD


Markers Diagnosis Assessment of Assessment of Predictor
Severity Treatment
Effects
FEV1 [% pred] NO YES ? YES ? YES
PaO2/PaCO2/SaO2 (%) NO YES [?] YES ///////////
Sputum volume/color YES YES YES ///////////
Imaging ? (differential NO NO NO
diagnosis)
Inflammatory markers /////////// /////////// /////////// YES ?
///////// indicates insufficient evidence

REFERENCES 10. Jones PW, Quirk FH, Baveystock CM, Littlejohns P. A self-
complete measure of health status for chronic airflow limi-
1. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd tation. The St. George's Respiratory Questionnaire. Am
SS. Global strategy for the diagnosis, management, and Rev Respir Dis 1992;145(6):1321-1327.
prevention of chronic obstructive pulmonary disease. 11. Dowson LJ, Newall C, Guest PJ, Hill SL, Stockley RA.
NHLBI/WHO Global Initiative for Chronic Obstructive Lung Exercise capacity predicts health status in alpha(1)-antit-
Disease (GOLD) Workshop summary. Am J Respir Crit rypsin deficiency. Am J Respir Crit Care Med
Care Med 2001;163(5):1256-1276. 2001;163(4):936-941.
2. Workshop Report: Global Strategy for Diagnosis, 12. Bestall JC, Paul EA, Garrod R, Garnham R, Jones PW,
Management and Prevention of COPD - Updated 2004. Wedzicha JA. Usefulness of the Medical Research Council
Available from http://www.goldcopd.org 2004. (MRC) dyspnoea scale as a measure of disability in
3. Jones PW, Agusti A. Markers and outcomes in the assess- patients with chronic obstructive pulmonary disease.
ment of chronic obstructive pulmonary disease. Eur Respir Thorax 1999;54(7):581-586.
J 2005;submitted. 13. Bowen JB, Votto JJ, Thrall RS, Haggerty MC, Stockdale-
4. Schols AM, Soeters PB, Dingemans AM, Mostert R, Woolley R, Bandyopadhyay T, ZuWallack RL. Functional
Frantzen PJ, Wouters EF. Prevalence and characteristics of status and survival following pulmonary rehabilitation.
nutritional depletion in patients with stable COPD eligible Chest 2000;118(3):697-703.
for pulmonary rehabilitation. Am Rev Respir Dis 14. Oga T, Nishimura K, Tsukino M, Sato S, Hajiro T. Analysis
1993;147(5):1151-1156. of the factors related to mortality in chronic obstructive pul-
5. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss monary disease: role of exercise capacity and health sta-
is a reversible factor in the prognosis of chronic obstructive tus. Am J Respir Crit Care Med 2003;167(4):544-549.
pulmonary disease. Am J Respir Crit Care Med 15. Rodriguez-Roisin R. Toward a consensus definition for
1998;157(6 Pt 1):1791-1797. COPD exacerbations. Chest 2000;117(5 Suppl 2):398S-401S.
6. Landbo C, Prescott E, Lange P, Vestbo J, Almdal TP. 16. Connors AF, Jr., Dawson NV, Thomas C, Harrell FE, Jr.,
Prognostic value of nutritional status in chronic obstructive Desbiens N, Fulkerson WJ, Kussin P, Bellamy P, Goldman
pulmonary disease. Am J Respir Crit Care Med L, Knaus WA. Outcomes following acute exacerbation of
1999;160(6):1856-1861. severe chronic obstructive lung disease. The SUPPORT
7. Mannino DM, Doherty DE, Sonia Buist A. Global Initiative investigators (Study to Understand Prognoses and
on Obstructive Lung Disease (GOLD) classification of lung Preferences for Outcomes and Risks of Treatments). Am J
disease and mortality: findings from the Atherosclerosis Respir Crit Care Med 1996;154(4 Pt 1):959-967.
Risk in Communities (ARIC) study. Respir Med 2005. 17. Breen D, Churches T, Hawker F, Torzillo PJ. Acute respira-
8. Vestbo J. The TORCH (towards a revolution in COPD tory failure secondary to chronic obstructive pulmonary
health) survival study protocol. Eur Respir J disease treated in the intensive care unit: a long term fol-
2004;24(2):206-210. low up study. Thorax 2002;57(1):29-33.
9. Meek PM, Lareau SC, Anderson D. Memory for symptoms 18. Agusti AG, Noguera A, Sauleda J, Sala E, Pons J,
in COPD patients: how accurate are their reports? Eur Busquets X. Systemic effects of chronic obstructive pul-
Respir J 2001;18(3):474-481. monary disease. Eur Respir J 2003;21(2):347-360.
19. Wouters EF. Management of severe COPD. Lancet
2004;364(9437):883-895.

OUTCOME AND MARKERS 113


GOLD_WR_05 8/18/05 12:56 PM Page 114

20. Ramsey SD, Sullivan SD. The burden of illness and eco- 35. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES,
nomic evaluation for COPD. Eur Respir J Suppl Harding GK, Nelson NA. Antibiotic therapy in exacerba-
2003;41:29s-35s. tions of chronic obstructive pulmonary disease. Ann Intern
21. Andersson F, Borg S, Jansson SA, Jonsson AC, Ericsson Med 1987;106(2):196-204.
A, Prutz C, Ronmark E, Lundback B. The costs of exacer- 36. Vestbo J. What is an exacerbation of COPD. Eur Respir J
bations in chronic obstructive pulmonary disease (COPD). 2004;13(88):6-13.
Respir Med 2002;96(9):700-708. 37. Bach PB, Brown C, Gelfand SE, McCrory DC.
22. Wouters EF. Economic analysis of the Confronting COPD Management of acute exacerbations of chronic obstructive
survey: an overview of results. Respir Med 2003;97 Suppl pulmonary disease: a summary and appraisal of published
C:S3-14. evidence. Ann Intern Med 2001;134(7):600-620.
23. Miravitlles M, Murio C, Guerrero T, Gisbert R. 38. Gross N. Outcome measures for COPD treatments, a criti-
Pharmacoeconomic evaluation of acute exacerbations of cal evaluation. J of COPD 2004;1:141-157.
chronic bronchitis and COPD. Chest 2002;121(5):1449-1455. 39. Quanjer PH, Tammeling GJ, Cotes JE, Pedersen OF,
24. Strassels SA, Smith DH, Sullivan SD, Mahajan PS. The Peslin R, Yernault JC. Lung volumes and forced ventilatory
costs of treating COPD in the United States. Chest flows. Report Working Party Standardization of Lung
2001;119(2):344-352. Function Tests, European Community for Steel and Coal.
25. Vollmer WM, Osborne ML, Buist AS. Uses and limitations Official Statement of the European Respiratory Society.
of mortality and health care utilization statistics in asthma Eur Respir J Suppl 1993;16:5-40.
research. Am J Respir Crit Care Med 1994;149(2 Pt 2): 40. Celli BR, MacNee W. Standards for the diagnosis and
S79-87; discussion S88-90. treatment of patients with COPD: a summary of the
26. Nishimura K, Izumi T, Tsukino M, Oga T. Dyspnea is a bet- ATS/ERS position paper. Eur Respir J 2004;23(6):932-946.
ter predictor of 5-year survival than airway obstruction in 41. Reid DW, Soltani A, Johns DP, Bish R, Williams TJ, Burns
patients with COPD. Chest 2002;121(5):1434-1440. GP, Walters EH. Bronchodilator reversibility in Australian
27. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca adults with chronic obstructive pulmonary disease. Intern
M, Mendez RA, Pinto Plata V, Cabral HJ. The body-mass Med J 2003;33(12):572-577.
index, airflow obstruction, dyspnea, and exercise capacity 42. Calverley PM, Burge PS, Spencer S, Anderson JA, Jones
index in chronic obstructive pulmonary disease. N Engl J PW. Bronchodilator reversibility testing in chronic obstruc-
Med 2004;350(10):1005-1012. tive pulmonary disease. Thorax 2003;58(8):659-664.
28. Casanova C, Cote C, de Torres JP, Aguirre-Jaime A, Marin 43. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson
JM, Pinto-Plata V, Celli BR. Inspiratory-to-total lung capaci- JA. Prednisolone response in patients with chronic obstruc-
ty ratio predicts mortality in patients with chronic obstruc- tive pulmonary disease: results from the ISOLDE study.
tive pulmonary disease. Am J Respir Crit Care Med Thorax 2003;58(8):654-658.
2005;171(6):591-597. 44. Traver GA, Cline MG, Burrows B. Predictors of mortality in
29. Jadad A, Rizo C, Cubillos P, Stahl E. Measuring symptom chronic obstructive pulmonary disease. A 15-year follow-up
response to pharmacological interventions in patients with study. Am Rev Respir Dis 1979;119(6):895-902.
COPD: a review of instruments used in clinical trials. Curr 45. Fletcher C, Peto R. The natural history of chronic airflow
Med Res Opin 2004;20(12):1993-2005. obstruction. Br Med J 1977;1(6077):1645-1648.
30. Jones PW, Kaplan RM. Methodological issues in evaluating 46. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey
measures of health as outcomes for COPD. Eur Respir J WC, Buist AS, Conway WA, Jr., Enright PL, Kanner RE,
Suppl 2003;41:13s-18s. O'Hara P, et al. Effects of smoking intervention and the use
31. Vestbo J, Prescott E, Lange P, Schnohr P, Jensen G. Vital of an inhaled anticholinergic bronchodilator on the rate of
prognosis after hospitalization for COPD: a study of a ran- decline of FEV1. The Lung Health Study. JAMA
dom population sample. Respir Med 1998;92(5):772-776. 1994;272(19):1497-1505.
32. Jones PW, Willits LR, Burge PS, Calverley PM. Disease 47. National Emphysema Treatment Trial Research Group.
severity and the effect of fluticasone propionate on chronic Patients at high risk of death after lung-volume-reduction
obstructive pulmonary disease exacerbations. Eur Respir J surgery. N Engl J Med 2001;345(15):1075-1083.
2003;21(1):68-73. 48. O'Donnell DE, Fluge T, Gerken F, Hamilton A, Webb K,
33. Fan VS, Curtis JR, Tu SP, McDonell MB, Fihn SD. Using Aguilaniu B, Make B, Magnussen H. Effects of tiotropium
quality of life to predict hospitalization and mortality in on lung hyperinflation, dyspnoea and exercise tolerance in
patients with obstructive lung diseases. Chest COPD. Eur Respir J 2004;23(6):832-840.
2002;122(2):429-436. 49. Anker SD, Negassa A, Coats AJ, Afzal R, Poole-Wilson
34. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson PA, Cohn JN, Yusuf S. Prognostic importance of weight
JA, Maslen TK. Randomised, double blind, placebo con- loss in chronic heart failure and the effect of treatment with
trolled study of fluticasone propionate in patients with mod- angiotensin-converting-enzyme inhibitors: an observational
erate to severe chronic obstructive pulmonary disease: the study. Lancet 2003;361(9363):1077-1083.
ISOLDE trial. Bmj 2000;320(7245):1297-1303.

114 OUTCOME AND MARKERS


GOLD_WR_05 8/18/05 12:56 PM Page 115

50. Agusti AG, Sauleda J, Miralles C, Gomez C, Togores B, 61. Barnes PJ, Shapiro SD, Pauwels RA. Chronic obstructive
Sala E, Batle S, Busquets X. Skeletal muscle apoptosis pulmonary disease: molecular and cellular mechanisms.
and weight loss in chronic obstructive pulmonary disease. Eur Respir J 2003;22(4):672-688.
Am J Respir Crit Care Med 2002;166(4):485-489. 62. Agusti AG. COPD, a multicomponent disease: implications
51. Miniati M, Filippi E, Falaschi F, Carrozzi L, Milne EN, for management. Respir Med 2005;99(6):670-682.
Sostman HD, Pistolesi M. Radiologic evaluation of emphy-
sema in patients with chronic obstructive pulmonary dis-
ease. Chest radiography versus high resolution computed
tomography. Am J Respir Crit Care Med 1995;151(5):1359-
1367.
52. Boschetto P, Miniati M, Miotto D, Braccioni F, De Rosa E,
Bononi I, Papi A, Saetta M, Fabbri LM, Mapp CE.
Predominant emphysema phenotype in chronic obstructive
pulmonary. Eur Respir J 2003;21(3):450-454.
53. Baldi S, Miniati M, Bellina CR, Battolla L, Catapano G,
Begliomini E, Giustini D, Giuntini C. Relationship between
extent of pulmonary emphysema by high-resolution com-
puted tomography and lung elastic recoil in patients with
chronic obstructive pulmonary disease. Am J Respir Crit
Care Med 2001;164(4):585-589.
54. Stolk J, Ng WH, Bakker ME, Reiber JH, Rabe KF, Putter H,
Stoel BC. Correlation between annual change in health
status and computer tomography derived lung density in
subjects with alpha1-antitrypsin deficiency. Thorax
2003;58(12):1027-1030.
55. Shaker SB, Stavngaard T, Stolk J, Stoel B, Dirksen A.
Alpha1-antitrypsin deficiency. 7: Computed tomographic
imaging in alpha1-antitrypsin deficiency. Thorax
2004;59(11):986-991.
56. Saetta M, Turato G, Maestrelli P, Mapp CE, Fabbri LM.
Cellular and structural bases of chronic obstructive pul-
monary disease. Am J Respir Crit Care Med
2001;163(6):1304-1309.
57. Sutherland ER, Martin RJ. Airway inflammation in chronic
obstructive pulmonary disease: comparisons with asthma.
J Allergy Clin Immunol 2003;112(5):819-827; quiz 828.
58. Pavord ID, Sterk PJ, Hargreave FE, Kips JC, Inman MD,
Louis R, Pizzichini MM, Bel EH, Pin I, Grootendorst DC,
Parameswaran K, Djukanovic R. Clinical applications of
assessment of airway inflammation using induced sputum.
Eur Respir J Suppl 2002;37:40s-43s.
59. Qiu Y, Zhu J, Bandi V, Atmar RL, Hattotuwa K, Guntupalli
KK, Jeffery PK. Biopsy neutrophilia, neutrophil chemokine
and receptor gene expression in severe exacerbations of
chronic obstructive pulmonary disease. Am J Respir Crit
Care Med 2003;168(8):968-975.
60. Gamble E, Grootendorst DC, Brightling CE, Troy S, Qiu Y,
Zhu J, Parker D, Matin D, Majumdar S, Vignola AM,
Kroegel C, Morell F, Hansel TT, Rennard SI, Compton C,
Amit O, Tat T, Edelson J, Pavord ID, Rabe KF, Barnes NC,
Jeffery PK. Antiinflammatory effects of the phosphodi-
esterase-4 inhibitor cilomilast (Ariflo) in chronic obstructive
pulmonary disease. Am J Respir Crit Care Med
2003;168(8):976-982.

OUTCOME AND MARKERS 115


GOLD_WR_05 8/18/05 12:56 PM Page 116

NOTES
GOLD_WR_05 8/18/05 12:56 PM Page 117

Printing and distribution has been made possible by educational grants from:

Visit the GOLD website at www.goldcopd.org

You might also like