Chronic Obstructive Pulmonary Disease (COPD)

Back

Practice Essentials

Chronic obstructive pulmonary disease (COPD) is estimated to affect 32 million persons in the United States and is the third leading cause of death in this country.[1] Patients typically have symptoms of chronic bronchitis and emphysema, but the classic triad also includes asthma or a combination of the above (see the image below).



View Image

Venn diagram of chronic obstructive pulmonary disease (COPD). Chronic obstructive lung disease is a disorder in which subsets of patients may have dom....

Chronic bronchitis is defined clinically as the presence of a chronic productive cough for 3 months during each of 2 consecutive years (other causes of cough being excluded).

Emphysema is defined pathologically as an abnormal, permanent enlargement of the air spaces distal to the terminal bronchioles, accompanied by destruction of their walls and without obvious fibrosis.

Signs and symptoms

Patients typically present with a combination of signs and symptoms of chronic bronchitis, emphysema, and reactive airway disease. Symptoms include the following:

While the sensitivity of physical examination in detecting mild-to-moderate COPD is relatively poor, the physical signs are quite specific and sensitive for severe disease. Findings in severe disease include the following:

Thoracic examination reveals the following:

Certain characteristics allow differentiation between disease that is predominantly chronic bronchitis and that which is predominantly emphysema. Chronic bronchitis characteristics include the following:

Emphysema characteristics include the following:

See Clinical Presentation for more detail.

Diagnosis

The formal diagnosis of COPD is made with spirometry; when the ratio of forced expiratory volume in 1 second over forced vital capacity (FEV1/FVC) is less than 70% of that predicted for a matched control, it is diagnostic for a significant obstructive defect. Criteria for assessing the severity of airflow obstruction (based on the percent predicted postbronchodilator FEV1) are as follows:

Arterial blood gas (ABG) findings are as follows:

In patients with emphysema, frontal and lateral chest radiographs reveal the following:

Advantages of high-resolution CT include the following:

Other tests are as follows:

See Workup for more detail.

Management

Smoking cessation continues to be the most important therapeutic intervention for COPD. Risk factor reduction (eg, influenza vaccine) is appropriate for all stages of COPD. Approaches to management by stage include the following:

Agents used include the following:

Pulmonary rehabilitation programs are typically multidisciplinary approaches that emphasize the following:

Indications for admission for acute exacerbations include the following:

See Treatment and Medication for more detail.

Background

Chronic obstructive pulmonary disease (COPD) is estimated to affect 32 million persons in the United States and is the third leading cause of death in the United States.[1] Patients typically have symptoms of chronic bronchitis and emphysema, but the classic triad also includes asthma (as seen in the image below). (See Clinical Presentation.)



View Image

Venn diagram of chronic obstructive pulmonary disease (COPD). Chronic obstructive lung disease is a disorder in which subsets of patients may have dom....

In Western Europe, Badham (1808) and Laennec (1827) made the classic descriptions of chronic bronchitis and emphysema in the early 19th century. A British medical textbook of the 1860s described the familiar clinical picture of chronic bronchitis as an advanced disease with repeated bronchial infections that ended in right-sided heart failure. Overall, this malady caused more than 5% of all deaths in the Middle Ages and earlier. The condition was most common among the poor; therefore, it was attributed to "bad" living.

Developments in the 20th century included the widespread use of spirometry (see Workup), recognition of airflow obstruction as a key factor in determining disability, and the improvement of pathologic methods to assess emphysema. Participants in the Ciba symposium of 1958 proposed definitions of chronic bronchitis and emphysema, incorporating the concept of airflow obstruction.

Chronic bronchitis is defined clinically as the presence of a chronic productive cough for 3 months during each of 2 consecutive years (other causes of cough being excluded). Emphysema, on the other hand, is defined pathologically as an abnormal, permanent enlargement of the air spaces distal to the terminal bronchioles, accompanied by destruction of their walls and without obvious fibrosis.

Airflow limitation in emphysema is due to loss of elastic recoil and decrease in airway tethering, whereas chronic bronchitis leads to narrowing of airway caliber and increase in airway resistance. Although some patients predominantly display signs of one of these diseases or the other, most fall somewhere in between the spectrum of these two conditions.

Past guidelines of COPD have been pessimistic at best, indicating that the disease process is irreversible and that therapy has little to offer. However, a more optimistic view has come to be widely accepted. The Global Initiative for Chronic Obstructive Lung Disease (GOLD) guidelines define COPD as a disease state characterized by airflow limitation that is not fully reversible, is usually progressive, and is associated with an abnormal inflammatory response of the lungs to inhaled noxious particles or gases,[4] (See Clinical Presentation.)

Oral and inhaled medications are used for patients with stable COPD to reduce dyspnea, improve exercise tolerance, and prevent complications. Most of the medications used in COPD treatment are directed at the potentially reversible mechanisms of airflow limitation. (See Medication.)

Pathophysiology

Pathologic changes in chronic obstructive pulmonary disease (COPD) occur in the large (central) airways, the small (peripheral) bronchioles, and the lung parenchyma. Most cases of COPD are the result of exposure to noxious stimuli, most often cigarette smoke. The normal inflammatory response is amplified in persons prone to COPD development. The pathogenic mechanisms are not clear but are most likely diverse. Increased numbers of activated polymorphonuclear leukocytes and macrophages release elastases in a manner that cannot be counteracted effectively by antiproteases, resulting in lung destruction.

The primary offender has been found to be human leukocyte elastase, with synergistic roles suggested for proteinase-3 and macrophage-derived matrix metalloproteinases (MMPs), cysteine proteinases, and a plasminogen activator. Additionally, increased oxidative stress caused by free radicals in cigarette smoke, the oxidants released by phagocytes, and polymorphonuclear leukocytes all may lead to apoptosis or necrosis of exposed cells. Accelerated aging and autoimmune mechanisms have also been proposed as having roles in the pathogenesis of COPD.[5, 6]

Cigarette smoke causes neutrophil influx, which is required for the secretion of MMPs; this suggests, therefore, that neutrophils and macrophages are required for the development of emphysema.

Studies have also shown that in addition to macrophages, T lymphocytes, particularly CD8+, play an important role in the pathogenesis of smoking-induced airflow limitation.

To support the inflammation hypothesis further, a stepwise increase in alveolar inflammation has been found in surgical specimens from patients without COPD versus patients with mild or severe emphysema. Indeed, mounting evidence supports the concept that dysregulation of apoptosis and defective clearance of apoptotic cells by macrophages play a prominent role in airway inflammation, particularly in emphysema.[7] Azithromycin (Zithromax) has been shown to improve this macrophage clearance function, providing a possible future treatment modality.[8]

In patients with stable COPD without known cardiovascular disease, there is a high prevalence of microalbuminuria, which is associated with hypoxemia independent of other risk factors.[9]

Chronic bronchitis

Mucous gland hyperplasia (as seen in the images below) is the histologic hallmark of chronic bronchitis. Airway structural changes include atrophy, focal squamous metaplasia, ciliary abnormalities, variable amounts of airway smooth muscle hyperplasia, inflammation, and bronchial wall thickening.



View Image

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells.



View Image

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells (high-powered vi....

Damage to the endothelium impairs the mucociliary response that clears bacteria and mucus. Inflammation and secretions provide the obstructive component of chronic bronchitis. Neutrophilia develops in the airway lumen, and neutrophilic infiltrates accumulate in the submucosa. The respiratory bronchioles display a mononuclear inflammatory process, lumen occlusion by mucus plugging, goblet cell metaplasia, smooth muscle hyperplasia, and distortion due to fibrosis. These changes, combined with loss of supporting alveolar attachments, cause airflow limitation by allowing airway walls to deform and narrow the airway lumen.

In contrast to emphysema, chronic bronchitis is associated with a relatively undamaged pulmonary capillary bed. The body responds by decreasing ventilation and increasing cardiac output. This V/Q mismatch results in rapid circulation in a poorly ventilated lung, leading to hypoxemia and polycythemia. Eventually, hypercapnia and respiratory acidosis develop, leading to pulmonary artery vasoconstriction and cor pulmonale. With the ensuing hypoxemia, polycythemia, and increased CO2 retention, these patients have signs of right heart failure and are known as "blue bloaters."

Emphysema

Emphysema is a pathologic diagnosis defined by permanent enlargement of airspaces distal to the terminal bronchioles. This leads to a dramatic decline in the alveolar surface area available for gas exchange. Furthermore, loss of alveoli leads to airflow limitation by 2 mechanisms. First, loss of the alveolar walls results in a decrease in elastic recoil, which leads to airflow limitation. Second, loss of the alveolar supporting structure leads to airway narrowing, which further limits airflow.

Emphysema has 3 morphologic patterns:

Centriacinar emphysema is characterized by focal destruction limited to the respiratory bronchioles and the central portions of the acini. This form of emphysema is associated with cigarette smoking and is typically most severe in the upper lobes.

Panacinar emphysema involves the entire alveolus distal to the terminal bronchiole. The panacinar type is typically most severe in the lower lung zones and generally develops in patients with homozygous alpha1-antitrypsin (AAT) deficiency.

Distal acinar emphysema, or paraseptal emphysema, is the least common form and involves distal airway structures, alveolar ducts, and sacs. This form of emphysema is localized to fibrous septa or to the pleura and leads to formation of bullae (as seen in the images below). The apical bullae may cause pneumothorax. Paraseptal emphysema is not associated with airflow obstruction.



View Image

Gross pathology of advanced emphysema. Large bullae are present on the surface of the lung.



View Image

Gross pathology of a patient with emphysema showing bullae on the surface.

The gradual destruction of alveolar septae (shown in the image below) and of the pulmonary capillary bed in emphysema leads to a decreased ability to oxygenate blood. The body compensates with lowered cardiac output and hyperventilation. This V/Q mismatch results in relatively limited blood flow through a fairly well oxygenated lung with normal blood gases and pressures in the lung, in contrast to the situation in chronic bronchitis. Because of low cardiac output, the rest of the body also suffers from tissue hypoxia and pulmonary cachexia. Eventually, these patients develop muscle wasting and weight loss and are identified as "pink puffers."



View Image

At high magnification, loss of alveolar walls and dilatation of airspaces in emphysema can be seen.

Emphysematous destruction and small airway inflammation

Emphysematous destruction and small airway inflammation often are found in combination in individual patients, leading to the spectrum that is known as COPD. When emphysema is moderate or severe, loss of elastic recoil, rather than bronchiolar disease, is the dominant mechanism of airflow limitation. By contrast, when emphysema is mild, bronchiolar abnormalities are most responsible for the majority of the deficit in lung function. Although airflow obstruction in emphysema is often irreversible, bronchoconstriction due to inflammation accounts for some reversibility. Airflow limitation is not the only pathophysiologic mechanism by which symptoms occur.

Dynamic hyperinflation

Lung volumes, particularly dynamic hyperinflation, have also been shown to play a crucial role in the development of dyspnea perceived during exercise. In fact, the improvement in exercise capacity brought about by several treatment modalities, including bronchodilators, oxygen therapy, lung volume reduction surgery (LVRS), and maneuvers learned in pulmonary rehabilitation, is more likely due to delaying dynamic hyperinflation rather than improving the degree of airflow obstruction.[10, 11, 12, 13, 14, 15, 16, 17] Additionally, hyperinflation (defined as the ratio of inspiratory capacity to total lung capacity [IC/TLC]) has been shown to predict survival better than forced expiratory volume in 1 second (FEV1).[7]

Etiology

Cigarette smoking

The primary cause of COPD is exposure to tobacco smoke. Overall, tobacco smoking accounts for as much as 90% of COPD risk.

Cigarette smoking induces macrophages to release neutrophil chemotactic factors and elastases, which lead to tissue destruction. Clinically significant COPD develops in 15% of cigarette smokers, although this number is believed to be an underestimate. Age of initiation of smoking, total pack-years, and current smoking status predict COPD mortality.

People who smoke have an increased annual decline in FEV1: the physiologic normal decline in FEV1 is estimated to be 20-30 ml/y, but the rate of decline in COPD patients is generally 60 ml/y or greater.

Secondhand smoke, or environmental tobacco smoke, increases the risk of respiratory infections, augments asthma symptoms, and causes a measurable reduction in pulmonary function.

A study by Nagelmann et al concluded that lung function deviation and lung structural changes are present in people who smoke cigarettes before the clinical signs of airway obstruction reveal them.[18] These changes can be detected by body plethysmography and diffusing capacity measurement with routine spirometry.

Environmental factors

COPD does occur in individuals who have never smoked.[19] Although the role of air pollution in the etiology of COPD is unclear, the effect is small when compared with that of cigarette smoking. In developing countries, the use of biomass fuels with indoor cooking and heating is likely to be a major contributor to the worldwide prevalence of COPD. Long-term exposure to traffic-related air pollution may be a factor in COPD in patients with diabetes and asthma.[20]

Airway hyperresponsiveness

Airway hyperresponsiveness (ie, Dutch hypothesis) stipulates that patients who have nonspecific airway hyperreactivity and who smoke are at increased risk of developing COPD with an accelerated decline in lung function. Nonspecific airway hyperreactivity is inversely related to FEV1 and may predict a decline in lung function.

The data regarding the possible role of airway hyperresponsiveness as a risk factor for the development of COPD in people who smoke are unclear. It is important to note, however, that 60% demonstrate bronchial hyperresponsiveness.[21] Moreover, bronchial hyperreactivity may result from airway inflammation observed with the development of smoking-related chronic bronchitis. This may contribute to airway remodeling, leading to a more fixed obstruction, as is seen in persons with COPD.

Alpha1-antitrypsin deficiency

Alpha1-antitrypsin (AAT) is a glycoprotein member of the serine protease inhibitor family that is synthesized in the liver and is secreted into the bloodstream. The main purpose of this 394-amino-acid, single-chain protein is to neutralize neutrophil elastase in the lung interstitium and to protect the lung parenchyma from elastolytic breakdown. Severe AAT deficiency predisposes to unopposed elastolysis with the clinical sequela of an early onset of panacinar emphysema. To see complete information on Alpha1-Antitrypsin Deficiency, please go to the main article by clicking here.

AAT deficiency is the only known genetic risk factor for developing COPD and accounts for less than 1% of all cases in the United States. Severe AAT deficiency leads to premature emphysema at an average age of 53 years for nonsmokers and 40 years for smokers.

Nearly 24 variants of the AAT molecule have been identified, and all are inherited as codominant alleles. The most common M allele (PiM) may be found in 90% of people, and homozygous (PiMM) phenotypes produce serum levels within the reference range. The homozygous PiZZ state is the most common deficiency state and accounts for 95% of people in the severely deficient category.

Intravenous drug use

Emphysema occurs in approximately 2% of persons who use intravenous (IV) drugs. This is attributed to pulmonary vascular damage that results from the insoluble filler (eg, cornstarch, cotton fibers, cellulose, talc) contained in methadone or methylphenidate.

The bullous cysts found in association with IV use of cocaine or heroin occur predominantly in the upper lobes. In contrast, methadone and methylphenidate injections are associated with basilar and panacinar emphysema.

Immunodeficiency syndromes

Human immunodeficiency virus (HIV) infection has been found to be an independent risk factor for COPD, even after controlling for confounding variables such as smoking, IV drug use, race, and age.[22]

Apical and cortical bullous lung damage occurs in patients who have autoimmune deficiency syndrome and Pneumocystis carinii infection. Reversible pneumatoceles are observed in 10-20% of patients with this infection.

Vasculitis syndrome

Hypocomplementemic vasculitis urticaria syndrome (HVUS) may be associated with obstructive lung disease. Other manifestations include angioedema, nondeforming arthritis, sinusitis, conjunctivitis, and pericarditis.

Connective tissue disorders

Cutis laxa is a disorder of elastin that is characterized most prominently by the appearance of premature aging. The disease usually is congenital, with various forms of inheritance (ie, dominant, recessive). Precocious emphysema has been described in association with cutis laxa as early as the neonatal period or infancy. The pathogenesis of this disorder includes a defect in the synthesis of elastin or tropoelastin.

Marfan syndrome is an autosomal dominant inherited disease of type I collagen characterized by abnormal length of the extremities, subluxation of the lenses, and cardiovascular abnormality. Pulmonary abnormalities, including emphysema, have been described in approximately 10% of patients.

Ehlers-Danlos syndrome refers to a group of inherited connective tissue disorders with manifestations that include hyperextensibility of the skin and joints, easy bruisability, and pseudotumors; it has also been associated with a higher prevalence of COPD.

Salla disease

Salla disease is an autosomal recessive storage disorder described in Scandinavia; the disease is characterized by intralysosomal accumulation of sialic acid in various tissues. The most important clinical manifestations are severe mental retardation, ataxia, and nystagmus. Precocious emphysema has been described and likely is secondary to impaired inhibitory activity of serum trypsin.

Epidemiology

The National Health Interview Survey reports the prevalence of emphysema at 18 cases per 1000 persons and chronic bronchitis at 34 cases per 1000 persons.[23] While the rate of emphysema has stayed largely unchanged since 2000, the rate of chronic bronchitis has decreased. Another study estimates a prevalence of 10.1% in the United States.[24] However, the exact prevalence of COPD in the United States is believed to be underestimated. This is largely due to the fact that it is an underdiagnosed (and undertreated) disease, because most patients do not present for medical care until the disease is in a late stage.

The exact prevalence of COPD worldwide is largely unknown, but estimates have varied from 7-19%. The Burden of Obstructive Lung Disease (BOLD) study found a global prevalence of 10.1%.[25] Men were found to have a pooled prevalence of 11.8% and women 8.5%. The numbers vary in different regions of the world. Cape Town, South Africa, has the highest prevalence, affecting 22.2% of men and 16.7% of women.

Hannover, Germany, on the other hand, has the lowest prevalence, of 8.6% for men and 3.7% for women. The differences can be explained in part by site and sex differences in the prevalence of smoking. As noted above, these reports are widely believed to be underestimates because COPD is known to be underdiagnosed and undertreated. Additionally, the prevalence in women is believed to be increasing.

Although current rates of COPD in men are higher than the rates in women, the rates in women have been increasing. COPD occurs predominantly in individuals older than age 40 years.

Severe, early onset disease likely represents a distinct genotype and is more commonly seen in females, African Americans, and those with a maternal family history of COPD.[26]

A study by Mintz et al estimated the prevalence of unidentified COPD.[27] Using the Lung Function Questionnaire (LFQ) and spirometry results, the study determined that approximately 1 in 5 patients (21%) aged 30 years or older with a history of smoking for 10 years or longer seen in a primary care center is likely to have COPD.

Prognosis

COPD is the third leading cause of death in the United States.[1] In terms of COPD as the underlying cause of death, absolute mortality rates for US patients aged 25 years or older (2005) were 77.3 deaths per 100,000 males and 56.0 deaths per 100,000 females, or 64.3 persons per 100,000 overall. Internationally, overall mortality rates from COPD vary markedly, from more than 400 deaths per 100,000 males aged 65-74 years in Romania to fewer than 100 deaths per 100,000 population in Japan.

The FEV1 was used to predict outcome in COPD until other factors were identified to play a role in determining the outcome of COPD patients. These discoveries resulted in the creation of the multidimensional BODE index (body mass index, obstruction [FEV1], dyspnea [modified Medical Research Council dyspnea scale], and exercise capacity [6MWD]).[28] This index was developed to assess an individual’s risk of death or hospitalization.

Prognosis is based on a point system, with all 4 factors used to determine the score, as follows:

The approximate 4-year survival based on the point system above is as follows:

The use of a clinical scoring system reinforces that determinants of prognosis in COPD remain multifactorial. Waschki et al argued that objective assessments of physical activity, including 6-minute walk test results, are best able to predict mortality.[29] However, additional socioeconomic factors also likely play a role in COPD prognosis; for example, a retrospective cohort study highlighted the increased risk of COPD-related mortality in patients who reside in isolated rural areas.[30]

A study by Sundh et al determined that the Clinical COPD Questionnaire (CCQ), which estimates quality of life in patients with COPD, is effective.[31] The CCQ identified that heart disease, depression, and underweight status are independently associated with lower health-related quality of life in patients with COPD.

In a multicenter, prospective, observational study of 201 consecutive patients with moderate-to-severe COPD, Martinez-Garcia et al reported that in addition to smoking, pulmonary hypertension, and declining lung function (known risk factors for mortality in patients with COPD),[32] bronchiectasis (which is common in patients with moderate-to-severe COPD) is independently associated with increased risk of all-cause mortality.[32, 33]

In this study, those who had bronchiectasis were found to be 2.5 times more likely to die than those who did not.[33] Bronchiectasis remained an independent factor after adjustment for dyspnea, partial pressure of oxygen, body mass index, presence of potentially pathogenic microorganisms in sputum, presence of daily sputum production, number of severe exacerbations and peripheral albumin, and ultrasensitive C-reactive protein concentrations.

Patient Education

It is important to educate the patient with COPD about the disease and to encourage his or her active participation in therapy. The 2 most essential points for the patient to understand are as follows:

For more information, see Emphysema.

History

Most patients with chronic obstructive pulmonary disease (COPD) seek medical attention late in the course of their disease. Patients often ignore the symptoms because they start gradually and progress over the course of years. Patients often modify their lifestyle to minimize dyspnea and ignore cough and sputum production. With retroactive questioning, a multiyear history can be elicited.

Patients typically present with a combination of signs and symptoms of chronic bronchitis, emphysema, and reactive airway disease. These include cough, worsening dyspnea, progressive exercise intolerance, sputum production, and alteration in mental status. Symptoms include the following:

Systemic manifestations (decreased fat-free mass, impaired systemic muscle function, osteoporosis, anemia, depression, pulmonary hypertension, cor pulmonale, left-sided heart failure

A productive cough or an acute chest illness is common. The cough usually is worse in the mornings and produces a small amount of colorless sputum.

Breathlessness is the most significant symptom, but it usually does not occur until the sixth decade of life (although it may occur much earlier). By the time the FEV1 has fallen to 50% of predicted, the patient is usually breathless upon minimal exertion. Despite the fact that FEV1 is the most common variable used to grade the severity of COPD, although it is not the best predictor of mortality.

Wheezing may occur in some patients, particularly during exertion and exacerbations.

The value of patient history and physical examination was addressed in the 2011 update to the American College of Physicians/American College of Chest Physicians/American Thoracic Society/European Respiratory Society (ACP/ACCP/ATS/ERS) guideline for diagnosis and management of stable COPD. According to the 2011 guideline, a history of more than 40 pack-years of smoking was the best single predictor of airflow obstruction; however, the most helpful information was provided by a combination of the following 3 signs[34] :

If all 3 signs are absent, airflow obstruction can be nearly ruled out.[34]

With disease progression, intervals between acute exacerbations become shorter, and each exacerbation may be more severe. The rate of COPD exacerbations appears to reflect an independent susceptibility phenotype.[35]

COPD is now known to be a disease with systemic manifestations, and the quantification of these manifestations has proved to be a better predictor of mortality than lung function alone. Many patients with COPD may have decreased fat-free mass, impaired systemic muscle function, osteoporosis, anemia, depression, pulmonary hypertension, cor pulmonale, and even left-sided heart failure. Depression is not uncommon in subjects with COPD.[36]

In a study by Spitzer et al in Germany, airflow limitation as measured by spirometry was significantly more common in adults with posttraumatic stress disorder than in controls. Results were adjusted for lifestyle, clinical, and sociodemographic factors.[37]

In addition, COPD appears to increase the risk for mild cognitive impairment (MCI). Investigators from the Mayo Clinic Study of Aging—a population-based, cross-sectional study of 1,927 participants—reported an association between COPD and an increased risk of having MCI, MCI subtypes, and memory loss in elderly patients.[38, 39] They also observed a dose-response relationship between COPD duration and an increased risk for cognitive problems.

The prevalence of MCI was significantly higher in patients with COPD (n = 288) (27%) than in those without COPD (15%), and there was a nearly twofold higher odds ratio (1.87) for MCI in patients with COPD. Moreover, the odds ratio increased from 1.6 in patients with COPD for 5 years or less to 2.1 in those who had COPD for longer than 5 years.[38, 39]

Some important clinical and historical differences may help distinguish between the types of COPD. Classic findings for patients with chronic bronchitis include productive cough with gradual progression to intermittent dyspnea; frequent and recurrent pulmonary infections; and progressive cardiac/respiratory failure with edema and weight gain. Classic findings for patients with emphysema include a long history of progressive dyspnea with late onset of nonproductive cough; occasional mucopurulent relapses; and eventual cachexia and respiratory failure.

Physical Examination

The sensitivity of a physical examination in detecting mild to moderate COPD is relatively poor; however, physical signs are quite specific and sensitive for severe disease. Patients with severe disease experience tachypnea and respiratory distress with simple activities.

The respiratory rate increases in proportion to disease severity. Use of accessory respiratory muscles and paradoxical indrawing of lower intercostal spaces is evident (known as the Hoover sign). In advanced disease, cyanosis, elevated jugular venous pulse (JVP), and peripheral edema can be observed.

Thoracic examination reveals the following:

In addition, coarse crackles beginning with inspiration may be heard.

Certain characteristics allow differentiation between disease that is predominantly chronic bronchitis and that which is predominantly emphysema.

Chronic bronchitis characteristics include the following:

Emphysema characteristics include the following:

Staging

The severity of airflow obstruction was the primary means of staging COPD until the American Thoracic Society (ATS) provided criteria for staging COPD based on the presence of obstruction (ratio of FEV1 to forced vital capacity [FEV1/FVC] < 70%) and its severity as measured by percent of predicted FEV1.

ATS and Global Initiative for Chronic Obstructive Lung Disease (GOLD) criteria for assessing the severity of airflow obstruction (based on the percent predicted postbronchodilator FEV1 when the FEV1/FVC is < 70%) are as follows:

However, these staging systems have limited utility in predicting mortality. The recognition that COPD is a systemic disease has helped in developing criteria that are better at predicting mortality than is assessment of airway obstruction alone. A widely used system for COPD prognosis is the BODE index (body mass index, obstruction [FEV1], dyspnea [modified Medical Research Council dyspnea scale], and exercise capacity [6MWD]).[28]

Approach Considerations

The defining feature of COPD is irreversible airflow limitation during forced expiration. This may result from a loss of elastic recoil due to lung tissue destruction or from an increase in the resistance of the conducting airways. The formal diagnosis of COPD is made with spirometry; when the ratio of forced expiratory volume in 1 second over forced vital capacity (FEV1/FVC) is less than 70% of that predicted for a matched control, it is diagnostic for a significant obstructive defect. Other studies, including laboratory studies and imaging, are particularly important during acute exacerbations of disease.

No blood-based biomarkers are accepted in COPD. However, a study by Sin et al investigated the use of serum pulmonary and activation-regulated chemokine (PARC/CCL-18) as a potential biomarker.[42] The study determined that PARC/CCL-18 levels are elevated in COPD and track clinical outcomes.

A large retrospective review from the United Kingdom found that in the 5 years preceding a diagnosis of COPD, primary care practitioners and specialists had missed opportunities to diagnose the disease in 85% of patients. Of the almost 39,000 patients in the study, 32,900 had presented with clinical or test findings consistent with early COPD. Missed opportunities included lower respiratory tract consultations resulting in a prescription for antibiotics or oral steroids and chest radiography not leading to a COPD diagnosis. Women were more likely to be underdiagnosed than men.[43]

Arterial Blood Gas Analysis

Arterial blood gas (ABG) analysis provides the best clues as to acuteness and severity of disease exacerbation.

Patients with mild COPD have mild to moderate hypoxemia without hypercapnia. As the disease progresses, hypoxemia worsens and hypercapnia may develop, with the latter commonly being observed as the FEV1 falls below 1 L/s or 30% of the predicted value. Lung mechanics and gas exchange worsen during acute exacerbations.

In general, renal compensation occurs even in chronic CO2 retainers (ie, bronchitics); thus, pH usually is near normal. Generally, consider any pH below 7.3 to be a sign of acute respiratory compromise.

Serum Chemistries

Patients with COPD tend to retain sodium. In addition, serum potassium should be monitored carefully, because diuretics, beta-adrenergic agonists, and theophylline act to lower potassium levels.

Beta-adrenergic agonists also increase renal excretion of serum calcium and magnesium, which may be important in the presence of hypokalemia.

Chronic respiratory acidosis leads to compensatory metabolic alkalosis. In the absence of blood gas measurements, bicarbonate levels are useful for following disease progression.

Alpha1-Antitrypsin

Measure alpha1-antitrypsin (AAT) in all patients younger than 40 years, in those with a family history of emphysema at an early age, or patients with emphysematous changes with no smoking history. The diagnosis of severe AAT deficiency is confirmed when the serum level falls below the protective threshold value of 11 mmol/L (ie, in the range of 3-7 mmol/L).

Specific phenotyping is reserved for patients in whom serum levels are 7-11 mmol/L or when genetic counseling or family analysis is needed.

Also see Alpha1-Antitrypsin Deficiency.

Sputum Evaluation

In persons with stable chronic bronchitis, the sputum is mucoid and macrophages are the predominant cells. With an exacerbation, sputum becomes purulent because of the presence of neutrophils. Although the quality of sputum can vary between patients in chronic stable disease, an increase in the quantity of sputum production is often a sign of an acute exacerbation.

A mixture of organisms often is visible with Gram stain (see Sputum Culture). The pathogens cultured most frequently during exacerbations are Streptococcus pneumoniae and Haemophilus influenzae.Moraxella catarrhalis is also a common organism, and Pseudomonas aeruginosa can be seen in patients with severe obstruction.

B-Type Natriuretic Peptide

Human B-type natriuretic peptide (BNP) binds to particulate guanylate cyclase receptors of vascular smooth muscle and endothelial cells. Binding to the receptors causes an increase in cyclic guanosine monophosphate (GMP), which serves as a secondary messenger to dilate veins and arteries. BNP is secreted by the ventricles of the heart when there is increased stretch of the myocytes (ie, in CHF).

By measuring BNP, it was thought that the ability to differentiate between CHF and COPD exacerbations would become much easier. However, clinical observation and research have shown that in cases of mild CHF exacerbation, differentiation between CHF and COPD is still not straightforward. A mild elevation of BNP must be taken in context with the overall clinical picture.

New biomarkers such as pro-BNP peptide assays are in development and may prove helpful in differentiating COPD from CHF exacerbations in the future.

Chest Radiography

As demonstrated in the images below, frontal and lateral chest radiographs of patients with emphysema reveal signs of hyperinflation, including flattening of the diaphragm, increased retrosternal air space, and a long, narrow heart shadow. Rapidly tapering vascular shadows accompanied by hyperlucency of the lungs are other signs of emphysema.

Chronic bronchitis is associated with increased bronchovascular markings and cardiomegaly.

With complicating pulmonary hypertension, the hilar vascular shadows are prominent, with possible right ventricular enlargement and opacity in the lower retrosternal air space.



View Image

Posteroanterior (PA) and lateral chest radiograph in a patient with severe chronic obstructive pulmonary disease (COPD). Hyperinflation, depressed dia....



View Image

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on lateral chest radiogr....



View Image

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on posteroanterior chest....

Computed Tomography

High-resolution CT (HRCT) scanning is more sensitive than standard chest radiography and is highly specific for diagnosing emphysema (outlined bullae are not always visible on a radiograph).

HRCT scanning may provide an adjunct means of diagnosing various forms of COPD (ie, lower lobe disease may suggest AAT deficiency) and may help the clinician to determine whether surgical intervention would benefit the patient. (See the CT image below.)



View Image

Severe bullous disease as seen on a computed tomography (CT) scan in a patient with chronic obstructive pulmonary disease (COPD).

Two-Dimensional Echocardiography

Many patients with long-standing COPD develop secondary pulmonary hypertension from chronic hypoxemia and vascular remodeling. This may result in eventual right-sided heart failure (cor pulmonale). However, even with severe COPD, the degree of pulmonary hypertension is usually only mild to moderate. Findings of severe pulmonary hypertension on echocardiogram or cardiac catheterization warrant further workup.

Two-dimensional echocardiography may be helpful as a screening tool to estimate pulmonary arterial systolic pressure and right ventricular systolic function, although formal cardiac catheterization is necessary to accurately confirm the diagnosis.

Pulmonary Function Tests

Pulmonary function tests are essential for the diagnosis and assessment of the severity of disease, and they are helpful in following its progress. (See the images below.) FEV1 is a reproducible test and is the most commonly used index of airflow obstruction.

In addition to the spirometry findings that define the disease, lung volume measurements often show an increase in total lung capacity, functional residual capacity, and residual volume. The vital capacity often decreases. Dynamic hyperinflation during exercise is now thought be a greater contributor to the sensation of dyspnea than airflow obstruction alone (as measured by FEV1).

As many as 30% of patients have an increase in FEV1 of 15% or more after inhalation of a bronchodilator. However, the absence of bronchodilator response does not justify withholding therapy.

Carbon monoxide diffusing capacity is decreased in proportion to the severity of emphysema.



View Image

Pressure volume curve comparing lungs with emphysema, lungs with restrictive disease, and normal lungs.



View Image

Flow volume curve of a patient with emphysema shows marked decrease in expiratory flow, hyperinflation, and air trapping (patient B) compared with a p....



View Image

Forced expiratory volume in 1 second (FEV1) can be used to evaluate the prognosis in patients with emphysema. The benefit of smoking cessation is show....

Using lung function thresholds of 80% predicted and fixed cut points to determine whether a test result is abnormal could result in the misdiagnosis of more than 20% of patients referred for pulmonary function testing. This misclassification can be avoided by using the lower limit of normal-based 95% confidence interval.[44]

Six-Minute Walking Distance

The distance walked in 6 minutes (6MWD) is a good predictor of all-cause and respiratory mortality in patients with moderate COPD.[2, 3] Patients with COPD who desaturate during the 6MWD have a higher mortality rate than do those who do not desaturate.

Consequently, this test is used as a part of the BODE index (body mass index, obstruction [FEV1], dyspnea [modified Medical Research Council dyspnea scale], and exercise capacity [6MWD]),[28] which was designed to help predict mortality in COPD patients.

Other Studies

Pulse oximetry does not offer as much information as arterial blood gas (ABG) analysis. However, when combined with clinical observation, this test can be a powerful tool for instant feedback on a patient's status.

Electrocardiography

Coexisting cardiac disease is highly likely in patients with COPD. Electrocardiography can be used in establishing that hypoxia is not resulting in cardiac ischemia and that the underlying cause of respiratory difficulty is not cardiac in nature.

Right-Sided Heart catheterization

If pulmonary hypertension is suspected based on clinical findings or on estimates from 2-dimensional echocardiography, then right-sided heart catheterization may be performed to measure pulmonary artery pressures directly and to gauge the response to vasodilators.

Hematocrit

Chronic hypoxemia may lead polycythemia. A hematocrit greater than 52% in men or 47% in women is indicative of polycythemia. Patients should be evaluated for hypoxemia at rest, with exertion, or during sleep. Correction of hypoxemia should reduce secondary polycythemia in patients who have quit smoking.

Approach Considerations

The goal of COPD management is to improve a patient’s functional status and quality of life by preserving optimal lung function, improving symptoms, and preventing the recurrence of exacerbations. Currently, no treatments aside from lung transplantation have been shown to significantly improve lung function or decrease mortality; however, oxygen therapy (when appropriate) and smoking cessation may reduce mortality. Once the diagnosis of COPD is established, it is important to educate the patient about the disease and to encourage his or her active participation in therapy.

Results of a randomized controlled trial showed that a comprehensive disease management strategy, which included a patient education session, a self-treatment plan for exacerbations, and a monthly follow-up call from a case manager, is associated with a lower hospitalization rate and fewer emergency department visits.[45] A study by Dewan et al determined that a multicomponent disease management program in patients with COPD was cost-effective, saving $593 per patient.[46]

Indications for intensive care admission are confusion, lethargy, respiratory muscle fatigue, worsening hypoxemia, and respiratory acidosis (pH < 7.30), as well as clinical concern for impending or active respiratory failure. (BiPAP can be done on the floor in some hospitals, including widely in the United Kingdom).

Oral and inhaled medications are used for patients with stable disease to reduce dyspnea and improve exercise tolerance. Most of the medications used are directed at the following 4 potentially reversible causes of airflow limitation in a disease state that has largely fixed obstruction:

Diet

Inadequate nutritional status associated with low body weight in patients with COPD is associated with impaired pulmonary status, reduced diaphragmatic mass, lower exercise capacity, and higher mortality rates. Nutritional support is an important part of comprehensive care in patients with COPD.

Bronchodilation

Bronchodilators are the backbone of any COPD treatment regimen. They work by dilating airways, thereby decreasing airflow resistance. This increases airflow and decreases dynamic hyperinflation. Lack of response in pulmonary function testing should not preclude their use. These drugs provide symptomatic relief but do not alter disease progression or decrease mortality.

Beta2 agonists and cholinergic/muscarinic antagonists

The initial choice of agent remains in debate. Historically, beta2 agonists were considered first line and anticholinergics were added as adjuncts. Not surprisingly, studies have shown combination therapy results in greater bronchodilator response and provides greater relief. Monotherapy with either agent and combination therapy with both are acceptable options. The adverse effect profile may help guide therapy. Generally, long-acting bronchodilators are more beneficial than short-acting ones.[2, 11, 16]

Beta2-agonist bronchodilators activate specific B2-adrenergic receptors on the surface of smooth muscle cells, which increases intracellular cyclic adenosine monophosphate (cAMP) and smooth muscle relaxation. Even patients who have no measurable increase in post-bronchodilator expiratory airflow may benefit from treatment with beta2 agonists. The inhaled route is preferred because it minimizes adverse systemic effects. The adverse effects are predictable and include tachycardia and tremors. Although rare, beta2 agonists may also precipitate a cardiac arrhythmia.

Anticholinergic drugs compete with acetylcholine for postganglionic muscarinic receptors, thereby inhibiting cholinergically mediated bronchomotor tone, resulting in bronchodilation. They block vagally mediated reflex arcs that cause bronchoconstriction. Clinical benefit is gained through a decrease in exercise-induced dynamic hyperinflation. These agents are poorly absorbed systemically and are relatively safe. Reported adverse effects include dry mouth, dry eyes, metallic taste, and prostatic symptoms.

In a study of men with COPD aged 66 years or older from Ontario, Canada, acute urinary retention (AUR) was found to be significantly more prevalent in users of inhaled anticholinergic medications (IACs) than in nonusers. The risk of AUR was higher in patients receiving both short-acting and long-acting IACs than in patients using a single IAC, and evidence of benign prostatic hyperplasia also increased the AUR risk.[47]

Inhaled delivery of medications is preferred over the oral route to help minimize potential adverse effects. Some patients may have difficulty achieving effective delivery of the medication using a metered-dose inhaler; use of a spacer or nebulizer may be beneficial in these patients.

The use of newly prescribed inhaled long-acting beta-agonist and long-acting anticholinergic drugs for COPD was associated with a 31% increased risk of experiencing a cardiovascular event in a recent nested case-control analysis of a retrospective cohort study. Both agents showed an increased risk of acute coronary syndrome and heart failure, but not arrhythmias or stroke. With both agents, the risk of events was highest within the first 2 or 3 weeks of initiating treatment. There was no significant difference in events between the 2 treatments.[48, 49]

In 2013, the results from the SPARK trial showed that in patients with severe COPD, a once-daily fixed-dose combination of indacaterol (a beta2 agonist) and glycopyrronium (a muscarinic antagonist) can improve lung functioning and reduce exacerbations, as compared with monotherapy with either glycopyrronium or tiotropium (both of which are muscarinic antagonists).[50]

Also in 2013, indacaterol and tiotropium were shown to be equally effective in a 52-week, randomized, blinded, parallel-group study consisting of more than 3400 patients with severe COPD who had had at least one exacerbation in the past year.[51, 52] Results from use of the 2 agents demonstrated similar improvements in baseline dyspnea and health status, with similar safety profiles.

Patients treated with indacaterol had a 29% higher exacerbation rate, but needed rescue medication less frequently than patients treated with tiotropium.[51, 52] In both groups, improvements were observed from baseline in trough FEV1 at week 12 (0.114 L with indacaterol and 0.126 L with tiotropium) and at week 52 (0.073 L with indacaterol and 0.092 L with tiotropium).[51, 52]

In 2015, glycopyrrolate was approved as a new respiratory inhalant dosage form. Glycopyrrolate is a long-acting muscarinic antagonist (LAMA) that produces bronchodilation by inhibiting acetylcholine’s effect on muscarinic receptors in the airway smooth muscle. It is available alone (Seebri Neohaler, encapsulated powder for inhalation; Lonhala Magnair, solution for nebulization) and in combination with indacaterol, a long-acting beta2-agonist (Utibron Neohaler).

Approval of indacaterol/glycopyrrolate inhaler was based the FLIGHT1 and FLIGHT2 12-week trials (n=2038) that measured lung function compared with its individual bronchodilator components, as well as placebo. Indacaterol/glycopyrrolate was statistically superior in terms of FEV1 area under the curve from 0-12 hours compared with its individual components (P< .001). Statistically and clinically meaningful improvements in St. George's Respiratory Questionnaire total score, transition dyspnea index total score, and reduction in rescue medication use were observed with indacaterol/glycopyrrolate compared with placebo (P< .001).[53]

Glycopyrrolate inhaled/formoterol (Bevespi Aerosphere) is a long-acting muscarinic antagonist (LAMA)/long-acting beta2-agonist (LABA) combination that was approved in April 2016. It is indicated for the long-term, maintenance treatment of airflow obstruction with COPD, including chronic bronchitis and/or emphysema. Approval is based on the PINNACLE trial program, which demonstrated that the glycopyrrolate/formoterol combination inhalant achieved statistically significant improvement in morning predose FEV1 at 24 weeks (P< .001) compared with the individual components of the combination and placebo.[54]

Umeclidinium bromide and vilanterol (Anoro Ellipta) is a LAMA and LABA inhalation powder approved by the FDA for long-term maintenance of COPD.[55, 56] Approval was based on a series of dose-ranging studies in more than 2400 patients—two 6-month, placebo-controlled efficacy and safety studies; two 6-month, active-controlled efficacy and safety studies; and a 12-month safety study.[56, 57]  This agent carries a boxed warning regarding use of LABAs and an increased risk of asthma-related death.[55, 56] Umeclidinium and vilanterol inhalation powder is not approved for asthma therapy, and it is not meant to be used as rescue therapy for sudden breathing problems (eg, acute bronchospasm). Serious adverse effects include paradoxical bronchospasm, cardiovascular effects, acute narrow-angle glaucoma, and worsening of urinary retention.[55, 56]

Olodaterol inhaled (Stirverdi Respimat) was approved by the FDA in July, 2014, for maintenance bronchodilator treatment in patients with COPD. Olodaterol is a long-acting beta2 agonist (LABA) that activates specific β2-adrenergic receptors on the surface of smooth muscle cells, which increases intracellular cAMP and smooth muscle relaxation. Approval was based on data from more than 3,000 patients with COPD studied over a 48-week period. Results demonstrated the long-term efficacy (eg, improved FEV1 at 0-3 hr and trough FEV1) and safety of once-daily olodaterol 5 mcg in patients with moderate to very severe COPD continuing with usual-care maintenance therapy.[58, 59]

Tiotropium

Although the results of the Understanding Potential Long Term Impacts on Function With Tiotropium (UPLIFT) trial did not show a change in the rate of decline of FEV1 or mortality when compared with placebo, it did show a significant reduction in the frequency of COPD exacerbations and hospitalizations and an improvement in quality of life.[60, 61, 62, 63]

Evidence is mounting of the efficacy of tiotropium over long-acting beta agonists. Two large, randomized trials have compared tiotropium, salmeterol (Serevent), and placebo.[61, 64] Both studies showed greater improvement in lung function, dyspnea, and quality of life in the tiotropium group versus the salmeterol group. A study by Brusasco et al also showed a delay in first exacerbations and fewer exacerbations per year in the tiotropium group.[64]

A 1-year, randomized, double-blind, double-dummy, parallel group study by Vogelmeier et al determined that tiotropium is more effective than salmeterol in preventing exacerbations in patients with moderate-to-very-severe COPD.[65]

Tiotropium is available in a capsule dosage form containing a dry powder for oral inhalation via the HandiHaler inhalation device. For adults, the contents of 1 capsule (18 mcg) are inhaled every day via the HandiHaler device. Contraindications, drug interactions, and adverse effects are similar to those of ipratropium.

A systematic review and meta-analysis by Singh et al of 5 randomized controlled trials of the Respimat tiotropium mist inhaler in patients with COPD found a 52% increased risk of mortality (all cause) versus placebo.[66] A randomized study comparing the Respimat mist inhaler to the HandiHaler inhalation device is ongoing as of August 2011.

Aclidinium

Aclidinium (Tudorza Pressair), a long-acting, antimuscarinic (M3) metered-dose inhaler was approved by the FDA in July 2012. Approval was based on randomized, placebo-controlled, clinical trials involving 1276 patients aged >40 years with COPD. The mean 12-week predose FEV1 improvements vs placebo were 0.12 L, 0.07 L, and 0.11 L (P< 0.001) in the trials, with a 24-week improvement of 0.13 L in the 6-month trial. Mean peak improvements in lung function (FEV1) assessed after the first dose were similar to those observed at week 12 in each study.[67]

Umeclidinium bromide

Umeclidinium bromide blocks the action of acetylcholine at muscarinic receptors in the bronchial airways (M3) by preventing increase in intracellular calcium concentration, leading to relaxation of airway smooth muscle. It is available in the United States as a combination inhaled powder with vilanterol (Anoro Ellipta), and is the first once-daily dual bronchodilator approved. It is also available as a single entity inhaler (Incruse Ellipta). Approval for the combination was based on 7 phase III trials including nearly 6,000 patients with COPD. Four 24-week primary efficacy studies (measuring improvement of trough FEV1) and a 52-week long-term safety study were the key studies. Two 12-week exercise/lung function studies provided supportive lung function data and contributed to safety data.[68, 69]

Revefenacin

Revefenacin is a once-daily, long-acting muscarinic antagonist that inhibits smooth-muscle M3 receptors in the airway, leading to bronchodilation. It is indicated for maintenance treatment of COPD. Revefenacin is administered by nebulizer using a mouthpiece. Approval was based on two 12-week, phase III trials including more than 1200 patients with COPD. Revefenacin demonstrated a significant improvement in baseline trough FEV1 compared with placebo in both trials.[70]

Phosphodiesterase inhibitors

Phosphodiesterase inhibitors increase intracellular cyclic adenosine monophosphate (cAMP) and result in bronchodilation. Additionally, they may improve diaphragm muscle contractility and stimulate the respiratory center.

Theophylline is a nonspecific phosphodiesterase inhibitor and is now limited to use as an adjunctive agent. Theophylline has a narrow therapeutic window with significant adverse effects, including anxiety, tremors, insomnia, nausea, cardiac arrhythmia (particularly multifocal atrial tachycardia), and seizures. It is reserved for patients with hard-to-control COPD or for individuals who are not able to use inhaled agents effectively.

Theophylline is metabolized primarily via the hepatic cytochrome P450 system, a process affected by age, cardiac status, and liver abnormalities. Serum levels of theophylline need to be monitored because of the potential for toxicity. The previously recommended target range of 15-20 mg/dL has now been reduced to 8-13 mg/dL.

Roflumilast (Daliresp) and cilomilast (Ariflo) are second-generation, selective phosphodiesterase-4 inhibitors. They cause a reduction of the inflammatory process (macrophages and CD8+ lymphocytes) in patients with COPD. Cilomilast is completely absorbed following oral administration and has a half-life of approximately 6.5 hours. A dose of 15 mg twice daily has been found to be clinically effective. Nausea, presumably of central origin, is the principal adverse reaction. The FDA advisory panel rejected approval of cilomilast in 2002.

Roflumilast was approved by the FDA in 2011 as a treatment to reduce the risk of COPD exacerbations in patients with severe COPD associated with chronic bronchitis and a history of exacerbations. In 2 randomized, double-blind, placebo-controlled, multicenter trials, increased FEV1 levels were found in patients who received roflumilast, and the rate of COPD exacerbations was reduced by 17% in these patients.[71]

Endogenous opioids

A study by Gifford et al found that the administration of endogenous opioids modulated the intensity and unpleasantness of breathlessness in patients with COPD.[72]

Beta-adrenergic antagonists (beta-blockers)

Cardiovascular disease is common in patients with COPD and is a leading cause of mortality; however, use of beta-blockers has been discouraged in these patients due to a perceived risk of bronchospasm and concern about inhibition of beta-agonist medication. However, a study by Short et al of 5977 patients in Scotland found that the addition of a cardioselective beta-blocker to established inhaled treatment did not appear to harm pulmonary function and did reduce COPD exacerbations, hospital admissions, and all cause mortality versus controls over a mean follow-up of 4.35 years.[73]

Smoking Cessation

Smoking cessation continues to be the most important therapeutic intervention for COPD. Most patients with COPD have a history of smoking or are currently smoking tobacco products. A smoking cessation plan is an essential part of a comprehensive management plan.

However, the success rates for cessation programs are low because of the addictive power of nicotine. These rates can also be negatively impacted by such factors as conditioned responses to smoking-associated stimuli, poor education, forceful promotional campaigns by the tobacco industry, and psychological problems, including depression. The process of smoking cessation typically requires multiple interventional approaches, including both pharmacologic and non-pharmacologic modalities, and will likely require multiple attempts to maintain success.

The transition from smoking to not smoking occurs in the following 5 stages:

Smoking intervention programs include self-help, group, health care provider delivered, workplace, and community programs.

Setting a quit date may be helpful. Physicians and other health care providers should participate in setting the target date and follow-up with respect to maintenance.

Successful cessation programs usually use the following resources and tools:

Mottillo et al reported meta-analysis results indicating that intensive behavioral interventions, including (but not limited to) individual counseling and telephone counseling, offer considerable benefit for increasing smoking abstinence.[74]

Supervised use of pharmacologic agents is an important adjunct to self-help and group smoking cessation programs.

Nicotine is the ingredient in cigarettes primarily responsible for tobacco addiction. Withdrawal from nicotine may cause unpleasant adverse effects, including anxiety, irritability, difficulty concentrating, anger, fatigue, drowsiness, depression, and sleep disruption. These effects usually occur during the first several weeks.

Nicotine replacement therapies after smoking cessation reduce withdrawal symptoms. If a smoker requires his or her first cigarette within 30 minutes of waking, the individual most likely is highly addicted and would benefit from nicotine replacement therapy.

Several nicotine replacement therapies are available.

Nicotine polacrilex (Nicorette, Nicorette Plus) is a chewing gum and has better quit rates than does counseling alone. Nicotine replacement therapy chewing pieces are marketed in 2 strengths (2 mg, 4 mg). An individual who smokes 1 pack per day should use 4-mg pieces. The 2-mg pieces are to be used by individuals who smoke less than 1 pack per day. Instruct patients to chew hourly and also to chew when needed for their initial cravings for 2 weeks. Gradually reduce the amount chewed over the next 3 months.

Transdermal nicotine patches are readily available for replacement therapy. Long-term success rates are 22-42%, compared with 2-25% for placebo. These agents are well tolerated, and the adverse effects are limited to local skin reactions. Nicotine replacement therapy patches are sold under the trade names NicoDerm, Nicotrol, and Habitrol. Each of these products is dosed with a scheduled graduated decrease in nicotine over 6-10 weeks.

The use of the antidepressant bupropion (Zyban) is also effective for smoking cessation. This nonnicotine aid to smoking cessation enhances central nervous system nonadrenergic function. One study demonstrated that 23% of patients sustained cessation at 1 year, compared with 12% who sustained cessation with the placebo. Bupropion may also be effective in patients who have not been able to quit smoking with nicotine replacement therapy.

Another drug used in smoking cessation is varenicline (Chantix). Varenicline is a partial agonist selective for alpha4, beta2 nicotinic acetylcholine receptors. Its action is thought to result from activity at a nicotinic receptor subtype, where its binding produces agonist activity while simultaneously preventing nicotine binding. Agonistic activity is significantly lower than nicotine.

Management of Inflammation

Inflammation plays a significant role in the pathogenesis of COPD. Systemic and inhaled corticosteroids attempt to temper this inflammation and positively alter the course of disease.

The use of systemic steroids in the treatment of acute exacerbations is widely accepted and recommended, given their high efficacy. A meta-analysis concluded that oral and parenteral corticosteroids significantly reduced treatment failure and the need for additional medical treatment and that they increased the rate of improvement in lung function and dyspnea over the first 72 hours.[75] Note that systemic steroids are not as effective in treating COPD exacerbations as they are in treating bronchial asthma exacerbations.

On the other hand, the use of oral steroids in persons with chronic stable COPD is widely discouraged, given their adverse effects, which include hypertension, glucose intolerance, osteoporosis, fractures, and cataracts. A Cochrane review showed no benefit at low-dose therapy and short-lived benefit with higher doses (>30 mg of prednisolone).[76]

Inhaled corticosteroids provide a more direct route of administration to the airways and, similar to other inhaled agents, are only minimally absorbed. Consequently, aside from the development of thrush, the systemic adverse effects of these medications at standard doses are negligible. Despite the theoretical benefit, the current consensus is that inhaled corticosteroids do not decrease the decline in FEV1, although they have been shown to decrease the frequency of exacerbations and improve quality of life for symptomatic patients with an FEV1 of less than 50%.[77]

Inhaled corticosteroids are not recommended as monotherapy and should be added to a regimen that already includes a long-acting bronchodilator. The Towards a Revolution in COPD Health (TORCH) trial showed that a combination of an inhaled corticosteroid and a long-acting beta agonist was more beneficial than inhaled corticosteroids alone.[78] These data suggest that in patients with COPD, inhaled corticosteroids should be used only in conjunction with a long-acting beta agonist.

However, patients treated with inhaled corticosteroids were noted to have an increased rate of pneumonia. The debate continues on the use of inhaled corticosteroids and the risk for pneumonia in patients with COPD. For example, no significant difference in pneumonia risk was found between patients who used inhaled budesonide and those who did not in a study by Sin et al. The authors analyzed data from 7 large clinical trials (n = 7042) of patients with stable COPD who used inhaled budesonide (n = 3801), with or without formoterol (Symbicort or Pulmicort, respectively) or a control regimen (placebo or formoterol alone [Oxis]). Increasing age and decreasing percent of predicted FEV1 were the only variables that were significantly associated with occurrence of pneumonia.[79]

Research has demonstrated that blood eosinophil count appears to be a promising biomarker of response to inhaled corticosteroids in patients with COPD. In one study, those patients with elevated eosinophil count reductions in exacerbations with combination inhaled corticosteroids and LABAs, compared with LABAs alone, were 24% in patients with baseline eosinophil counts of greater than or equal to 2% to less than 4%, 32% for those with counts of 4% to less than 6%, and 42% for those with eosinophil counts of greater than or equal to 6%.[80] Similar results were found in two other studies.[81, 82]

Despite the possible increased risk of pneumonia associated with inhaled corticosteroids, a retrospective cohort study showed that in patients with COPD hospitalized with pneumonia, prior use of inhaled corticosteroids was actually associated with decreased mortality and less mechanical ventilation.[83] Therefore the benefit of inhaled corticosteroids in selected patients will likely continue to outweigh the risks.

Intravenous steroids are often used in high doses for acute exacerbations in the inpatient setting; recent research suggests that there is likely no benefit of IV over oral steroid formulations in acute exacerbations, and thus IV steroids should be reserved only for those patients unable to tolerate oral intake.

Nonsteroidal anti-inflammatory medications have not been shown conclusively to have any benefit in COPD. No response has been shown to medications targeting interleukin-8 and tumor necrosis factor-alpha. Leukotriene inhibitors commonly used in asthma have also not proven to be beneficial in COPD.

However, macrolide antibiotics have been shown to have anti-inflammatory effects in the airways of COPD patients. More specifically, azithromycin has been shown to improve the phagocytic function of pulmonary macrophages and to be a potent anti-inflammatory.[8]

Azithromycin is used clinically for its anti-inflammatory effects in patients with cystic fibrosis and in lung transplantation patients with chronic rejection. Furthermore, one study showed that erythromycin reduced the frequency of exacerbations in 109 patients with COPD treated over 12 months.[84] A subsequent larger randomized controlled trial of 1142 patients showed a slight decrease in COPD exacerbations for patients given azithromycin over 1 year compared with placebo.[85] However, this study also noted an increase in hearing decrements in the patients receiving azithromycin (25% in the treatment group compared with 20% in the placebo group; P =0.04). The noted side effect combined with the concern for breeding antimicrobial resistance continues to prevent widespread use of azithromycin for prevention of COPD exacerbations.

Management of Infection

In patients with COPD, chronic infection or colonization of the lower airways is common from S pneumoniae, H influenzae, and M catarrhalis. In patients with chronic severe airway obstruction, P aeruginosa infection may also be prevalent. The use of antibiotics for the treatment of acute exacerbations is well supported.[23] Patients who benefited most from antibiotic therapy were those with exacerbations that were characterized by at least 2 of the following: increases in dyspnea, sputum production, and sputum purulence (The Winnipeg criteria). No evidence supports the continuous or prophylactic use of antibiotics to prevent exacerbations.

Empiric antimicrobial therapy is recommended in patients with an acute exacerbation (as evidenced by an increase in baseline dyspnea and/or a change in the quantity or quality of cough) and evidence of an infectious process, such as fever, leukocytosis, or an infiltrate on chest radiograph. The antibiotic choice must be comprehensive and should cover all likely pathogens in the context of the clinical setting and local resistance patterns.[4]

In a study by Daniels et al, the addition to doxycycline to corticosteroids was found to somewhat improve treatment for acute exacerbation of COPD (AECOPD). The investigators conducted a randomized, placebo-controlled trial that compared the addition of doxycycline to corticosteroids on clinical outcome in patients hospitalized with AECOPD. In addition to clinical outcome, other parameters were measured, including microbiological outcome, lung function, and systemic inflammation. The 223 patients enrolled in the study represented 265 COPD exacerbations. In addition to systemic corticosteroids, patients received either doxycycline at 200 mg or placebo for 7 days.

Results at 30 days were similar between the 2 groups. At 10 days, the doxycycline group showed superiority in clinical success compared with placebo in the intention-to-treat arm but not in the per-protocol arm. Also at day 10, doxycycline was superior for clinical cure, microbiological outcome, use of open label antibiotics, and symptoms.[86]

Management of Sputum Viscosity and Secretion Clearance

Mucolytic agents reduce sputum viscosity and improve secretion clearance. Viscous lung secretions in patients with COPD consist of mucus-derived glycoproteins and leukocyte-derived DNA.

The oral agent N -acetylcysteine has antioxidant and mucokinetic properties and is used to treat patients with COPD. However, the efficacy of mucolytic agents in the treatment of COPD remains controversial. Although they have been shown to decrease cough and chest discomfort, they have not been shown to improve dyspnea or lung function, and they have also been shown to elicit bronchospasm. When used as an inhalational therapy, N -acetylcysteine should be administered along with a bronchodilator such as albuterol in order to counteract potential induction of bronchospasm.

PPIs for Exacerbations and the Common Cold

In patients with COPD, the addition of a proton pump inhibitor (PPI) to conventional therapy may significantly decrease COPD exacerbations but not the incidence of the common cold, according to a study by Sasaki et al.[87] The investigators conducted a randomized, observer-blind, controlled trial to determine whether PPIs reduce the incidence of common colds in patients with COPD. Patients (n = 100) were assigned to conventional therapy (control group) or conventional therapy plus PPI (lansoprazole [Prevacid] 15 mg/d).

In the study by Sasaki et al, the frequency of common colds and COPD exacerbations was measured, and it was determined that the number of exacerbations per person over 12 months was significantly lower in the PPI group than in the control group. However, no significant difference in the frequency of common colds was observed between the 2 groups. The authors concluded that although the patients who took lansoprazole showed a significant decrease in COPD exacerbations, more definitive clinical trials are required.[87]

Oxygen Therapy and Hypoxemia

COPD is commonly associated with progressive hypoxemia. Oxygen administration reduces mortality rates in patients with advanced COPD because of the favorable effects on pulmonary hemodynamics.

Long-term oxygen therapy improves survival 2-fold or more in hypoxemic patients with COPD, according to 2 landmark trials, the British Medical Research Council (MRC) study and the US National Heart, Lung and Blood Institute’s Nocturnal Oxygen Therapy Trial (NOTT). Hypoxemia is defined as PaO2 (partial pressure of oxygen in arterial blood) of less than 55 mm Hg or oxygen saturation of less than 90%. Oxygen was used for 15-19 hours per day.[88, 89, 90]

Therefore, specialists recommend long-term oxygen therapy for patients with a PaO2 of less than 55 mm Hg, a PaO2 of less than 59 mm Hg with evidence of polycythemia, or cor pulmonale. Reevaluate these patients 1-3 months after initiating therapy, because some patients may not require long-term oxygen.

Many patients with COPD who are not hypoxemic at rest worsen during exertion. Home supplemental oxygen commonly is prescribed for these patients. Oxygen supplementation during exercise can prevent increases in pulmonary artery pressure, reduce dyspnea, and improve exercise tolerance.

In a 2008 study, however, patients with COPD-related hypoxemia and exertional desaturation who completed a program of pulmonary rehabilitation failed to show any benefit in domestic activity, health-related quality of life, or time spent outside of the home when compared with those who received placebo.[91] Hence, the benefits of home ambulatory oxygen for this subset of patients remain controversial.

Oxygen therapy generally is safe. Oxygen toxicity from high inspired concentrations (>60%) is well recognized. Little is known about the long-term effects of low-flow oxygen. However, the increased survival and quality-of-life benefits of long-term oxygen therapy outweigh the possible risks of oxygen toxicity.

Carbon dioxide retention from depression of hypoxic drive has been overemphasized. Despite the widely held belief that too much oxygen causes significant respiratory depression, multiple studies in the literature dispute this view. With administration of oxygen, PaCO2 rises, but not in proportion to the very minor changes in respiratory drive. Carbon dioxide retention is more likely a consequence of ventilation-perfusion mismatching rather than respiratory center depression. While this complication is not common, it is best avoided by titrating oxygen delivery to maintain the PaO2 at 60-65 mm Hg.

The major physical hazards of oxygen therapy are fires or explosions. Patients, family, and other caregivers must be warned not to smoke. Overall, major accidents are rare and can be avoided by good patient and family training.

The continuous-flow nasal cannula is the standard means of oxygen delivery for the stable hypoxemic patient. It is simple, reliable, and generally well tolerated. Each liter of oxygen flow adds 3-4% to the fraction of inspired oxygen (FiO2). Nasal oxygen delivery also is beneficial for most mouth-breathing patients. Humidification generally is not necessary when the patient receives oxygen by nasal cannula at flows of less than 5 L/min. (See the images below.)



View Image

Oxygen therapy via nasal cannula.



View Image

Home supplemental oxygen.

Oxygen-conserving devices function by delivering all of the supplemental oxygen during early inhalation. These devices improve the portability of oxygen therapy and may reduce overall costs. Three distinct oxygen-conserving devices are available: reservoir cannulas, demand-pulse delivery devices, and transtracheal oxygen delivery. Transtracheal oxygen delivery involves the insertion of a catheter percutaneously between the second and third tracheal interspace. Transtracheal oxygen delivery is invasive and requires special training for the physician, the patient, and the caregiver. The procedure has risks as well as medical benefits but has limited application.

NIPPV for hypercapneic respiratory failure

Noninvasive positive-pressure ventilation (NIPPV), as the name suggests, allows the delivery of positive-pressure ventilation without the use of an endotracheal tube. In place of the tube is a tight-fitting nasal or facial mask that is attached to a continuous positive airway pressure (CPAP) or a bilevel positive airway pressure (BiPAP) machine (as seen below). The positive pressure is beneficial in hypercapneic respiratory failure by decreasing the work of breathing, allowing a larger tidal volume for a given respiratory effort, and hence improving alveolar ventilation.[92]



View Image

Bilevel positive airway pressure (BiPAP).

NIPPV has been shown to provide significant benefits in selected patients with acute hypercapneic respiratory failure due to COPD, including a reduction in the need for endotracheal intubation, reduced hospital stay, and a mortality benefit.[93, 94] This modality should not be used in patients who are unable to protect their airway, are hemodynamically unstable, have significant secretions, are uncooperative, or have an Acute Physiology and Chronic Health Evaluation (APACHE) score of greater than 29.[95]

Another study suggests that in patients with chronic hypercapneic respiratory failure who are undergoing pulmonary rehabilitation, nocturnal NIPPV may improve quality of life, daytime PaCO2, and exercise tolerance.[88]

Vaccination to Reduce Infections

Infections can lead to COPD exacerbations. Vaccinations are a safe and effective modality to reduce infections in susceptible COPD patients. The pneumococcal vaccine should be offered to all patients older than 65 years or to patients of any age who have an FEV1 of less than 40% of predicted. The influenza vaccine should be given annually to all COPD patients.

Alpha1-Antitrypsin Deficiency Treatment

The treatment strategies for AAT deficiency involve reducing the neutrophil elastase burden, primarily by smoking cessation, and augmenting the levels of AAT. Available augmentation strategies include pharmacologic attempts to increase endogenous production of AAT by the liver (ie, danazol, tamoxifen) and administration of purified AAT by periodic intravenous infusion or by inhalation. Tamoxifen can increase endogenous production of AAT to a limited extent, so this may be beneficial in persons with the PISZ phenotype.

Intravenous AAT augmentation therapy is the only available approach that can increase serum levels to greater than 11 mmol/L, the protective threshold. Studies show that the infusions can maintain levels of more than 11 mmol/L, and replacement is administered weekly (60 mg/kg), biweekly (120 mg/kg), or monthly (250 mg/kg).

Purified AAT is currently available in 3 formulations, with trade names of Prolastin, Zemaira, and Aralast, all of which are derived from donor blood. Synthetic formulations of AAT are under development but not yet available. The ability of intravenous AAT augmentation to alter the clinical course of patients with AAT deficiency has not been demonstrated. Uncontrolled observations of patients suggest that the FEV1 may fall at a slower rate in patients who receive AAT replacement.[96]

Inpatient Care

Treatment of acute exacerbation of COPD

Acute exacerbations (AE) of COPDs are a major reason for hospital admission in the United States, although mild episodes may be treated in an outpatient setting. Indications for admission include failure of outpatient treatment, marked increase in dyspnea, altered mental status, increase in hypoxemia or hypercapnia, and inability to tolerate oral medications such as antibiotics or steroids. Care must be taken to evaluate for other conditions that may mimic AECOPD.

Patients with COPD are susceptible to many insults that can lead rapidly to an acute deterioration superimposed on chronic disease. AECOPD—an important, but occasionally overlooked, parameter—is defined as worsening of cough, increase in sputum production, change in sputum quality, and increase in dyspnea.

AECOPDs are very common, affecting about 20% of patients with moderate to severe COPD (1.3 events per year in patients with 40-45% of predicted FEV1). Quick and accurate recognition of these patients, along with aggressive and prompt intervention, may be the only action that prevents frank respiratory failure. Care must also be taken to evaluate for other conditions that may mimic acute exacerbations of COPD. 

AECOPDs occur in clusters, not at random, as previously thought, according to an analysis by Hurst et al.[97] Their study showed that patients with an AECOPD are at an increased risk of another attack in the 8 weeks following the initial episode. Close follow-up during this “brittle” period may lead to earlier treatment and better clinical outcomes.

A stepwise approach to drug therapy is recommended that takes into consideration the causes and complications related to the exacerbation, the degree of reversible bronchospasm, recent drug use, and contraindications to treatment. Sedation and pain management must be provided to ensure patient comfort and safety, despite a potential for respiratory depression.

Assisted ventilation

Progressive airflow obstruction may impair oxygenation and/or ventilation to the degree that the patient requires assisted ventilation. The general guidelines indicate that the ideal time to initiate ventilatory support are (1) when patients have experienced progressive worsening of respiratory acidosis and/or altered mental status and (2) when clinically significant hypoxemia exists despite supplemental oxygen.

Patients may be treated with noninvasive mask ventilation or with translaryngeal intubation and mechanical ventilation.

Bullectomy

The bullae in patients with emphysema generally range from 1-4 cm in diameter; on occasion, bullae can occupy more than 33% of the hemithorax (ie, giant bullae). Giant bullae may compress adjacent lung tissue, thereby reducing blood flow and ventilation to healthier tissue.

Removal of these bullae, a standard treatment in selected patients for many years, may result in the expansion of compressed lung tissue and improved function. Patients who are symptomatic and have an FEV1 of less than 50% of the predicted value have a better outcome after bullectomy.

Bullectomy is performed through a midline sternotomy or a lateral incision, or by video-assisted thoracoscopy. Postoperative bronchopleural air leak is the major potential complication.

Lung Volume Reduction Surgery

Nearly 40 years ago, Brantigan et al first reported resectional surgery for diffuse emphysema in 33 patients. They resected 20-30% of each lung that appeared most diseased. The investigators hypothesized that removal of a portion of the emphysematous lung would increase radial traction on the airways in the remaining lung, thereby reducing symptoms by improving expiratory airflow and mechanical function.

Lung volume reduction surgery is carried out using a midline sternotomy with stapling of the lung margins. Surgeons generally resect 20-30% of each lung from the upper zones. The lung volume reduction procedure has a mortality rate of 0-18%. Several complications, including pneumonia and prolonged air leaks, have been observed.

Several studies, including the large, multicenter National Emphysema Treatment Trial (NETT), have demonstrated significant benefit in spirometry, exercise tolerance, dyspnea, health-related quality of life, and mortality in selected patients after LVRS.[98]

Those who benefit most are patients with heterogeneous (upper lobe) disease and a low exercise capacity despite optimal medical therapy and cardiopulmonary rehabilitation. Patients with an FEV1 of less than 20% of predicted and either homogenous disease or DLCO (diffusing capacity of lung for carbon monoxide) of less than 20% of predicted are considered high risk for this procedure.

Lung Transplantation

Lung transplantation is performed only at select tertiary care centers around the world. Patients with COPD are the largest single category of patients who undergo the procedure.

When evaluating a potential candidate, several factors need be taken into account, including symptomatology, comorbid conditions, and projected survival without transplantation (eg, BODE index >5). Generally speaking, most centers set an age limit of 65 years.

The mean survival after lung transplantation is 5 years. The survival at 1 year is 80-90%.[99, 100] Whether this procedure has any effect on the survival of patients with COPD is controversial; the main purpose of lung transplantation is to improve symptomatology and quality of life.

Long-term Monitoring

Follow-up

Disposition after an AECOPD depends on the clinical picture for each patient more than on any single laboratory value or test. In general, the longer the exacerbation, the more airway edema and debris are present. Nearly all discharged patients should receive a short steroid burst and an increase in the frequency of inhaler therapy. Close follow-up should be arranged with the patient's regular care provider. Other therapies should be considered on a case-by-case basis.

Additional follow-up recommendations are as follows:

Pulmonary rehabilitation

Many patients with COPD are unable to enjoy life to the fullest because of shortness of breath, physical limitations, and inactivity. Pulmonary rehabilitation encompasses an array of therapeutic modalities designed to improve the patient's quality of life by decreasing airflow limitation, preventing secondary medical complications, and alleviating respiratory symptoms. The 3 major goals of the comprehensive management of COPD are the following:

Successful implementation of a pulmonary rehabilitation program usually requires a team approach, with individual components provided by healthcare professionals who have experience in managing COPD (eg, physician, dietitian, nurse, respiratory therapist, exercise physiologist, physical therapist, occupational therapist, recreational therapist, cardiorespiratory technician, pharmacist, psychosocial professionals). This multidisciplinary approach emphasizes the following:

As a result of rehabilitation, improvements have been shown in objective measures of patients’ quality of life, well-being, and health status, including reduction in respiratory symptoms, increases in exercise tolerance and functional activities (eg, walking), reduction in anxiety and depression, and increases in feelings of control and self-esteem.

An observational study has shown that pulmonary rehabilitation also improves the BODE index score in patients with COPD and is associated with better outcomes.[101] Quality of life improvements after pulmonary rehabilitation can also be measured using the COPD Assessment Test (CAT), an 8-question patient-completed instrument.[102] In addition, pulmonary rehabilitation results in substantial savings in health care costs by reducing hospitalizations and the use of medical resources.

Pulmonary rehabilitation programs usually are conducted in an outpatient setting. A rehabilitation program may include a number of components and should be tailored to the needs of the individual patient. Provide all patients who complete the program with guidelines for continuing at home.

Education is key to comprehensive pulmonary rehabilitation. The educational component prepares the patient and family to be actively involved in providing care. This reliance on patients to assume charge of their care is known as collaborative self-management.

Exercise training is a mandatory component of pulmonary rehabilitation. Patients with COPD should perform aerobic lower extremity endurance exercises regularly to enhance performance of daily activities and reduce dyspnea. Upper extremity exercise training improves dyspnea and allows increased activities of daily living requiring the use of upper extremities.

Breathing retraining techniques (eg, diaphragmatic, pursed lip breathing) may improve the ventilatory pattern and prevent dynamic airway compression.



View Image

Pulmonary rehabilitation.

Special concerns

Many commercial airplanes fly at altitudes of 30,000-40,000 feet. However, the cabin is pressurized to an altitude of 5000-8000 feet. At these altitudes, atmospheric partial pressure of oxygen (PO2) is 132-109 mm Hg, compared with 159 mm Hg at sea level.

Acute reduction in PO2 stimulates peripheral chemoreceptors, which results in hyperventilation. When determining whether or not a COPD patient can safely fly, the clinician should first use a prediction equation to determine whether the patient will become hypoxemic at high altitudes. A prediction equation used to estimate PaO2 at 8000 feet (2440 m) is as follows:

PaO2 = 22.8 - 2.74X + 0.68Y

(X = altitude; Y = arterial PO2 at sea level)

A predicted PaO2 of 50 mm Hg or less at an altitude of 8000 feet is an indication for supplemental oxygen. Arrange supplemental oxygen prior to the flight directly through the airline or through the airline agent (at an extra expense). If there is any question regarding the calculation, many pulmonary physiology labs can also perform altitude simulation tests to confirm or refute the need for in-flight oxygen.

Patients with COPD may develop substantial decreases in nocturnal PaO2 during all phases of sleep, but particularly during the rapid eye movement phase. These episodes are associated initially with a rise in pulmonary arterial pressures and a disturbance in sleep architecture, but they may develop into pulmonary arterial hypertension and cor pulmonale if hypoxemia remains untreated.

Prescribe oxygen for patients who have daytime PaO2 greater than 60 mm Hg but who demonstrate substantial nocturnal hypoxemia.

End-of-Life Care

COPD is a chronic disease that is preventable and treatable but largely incurable. Given the progressive nature of the disease, as well as the morbidity and mortality associated with it, clinicians should always include end-of-life care discussions in their visits with patients. These discussions should focus on palliative efforts to improve quality of life, as well as assistance with advanced directives, advanced care planning, and referrals for hospice and home care when needed.

Guidelines Summary

Guidelines related to chronic obstructive pulmonary disease (COPD) screening have been issued by the US Preventive Services Task Force (USPSTF).[103] Additionally, this organization published guidelines on preventing tobacco use and smoking cessation.[104]

Other guidelines include general COPD management guidelines (eg, GOLD guidelines)[4, 105] and US Department of Veterans Affairs/Department of Defense (VA/DoD) guidelines.[106] Joint guidelines for stable COPD management have been issued by the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society.[34]

Finally, the Canadian Critical Care Trials Group and the Canadian Critical Care Society Noninvasive Ventilation Guidelines Group issued guidelines on noninvasive positive-pressure ventilation (NIPPV),[107] and the American College of Chest Physicians and Canadian Thoracic Society released guidelines on COPD exacerbation prevention.[108]

Screening Guidelines

In 2016, the USPSTF retained its 2008 recommendation against screening for COPD in individuals who do not have related symptoms.[103] The task force found inadequate evidence in support of the value of questionnaires or spirometry in improving health outcomes (quality of life, morbidity, or mortality) of asymptomatic individuals. It found the screening procedures not overtly harmful, but costly in terms of time and expense. Moreover, the task force concluded that because screening and available drug treatments do not alter the course of the disease, screening offers no net benefit. This recommendation does not apply to adults who present with symptoms, such as chronic cough or dyspnea.

Tobacco Use Guidelines

According to the USPSTF guidelines on preventing tobacco use and tobacco-caused disease, clinicians should ask all adult patients about their use of tobacco products and provide cessation interventions to current users.[104] The guideline engages a "5-A" approach to counseling that includes the following:

Brief behavioral counseling (< 10 min) and pharmacotherapy are each effective alone—although they are most effective when used together. The USPSTF also advises clinicians to ask all pregnant women, regardless of age, about tobacco use. Those who currently smoke should receive pregnancy-tailored counseling supplemented with self-help materials.

Management Guidelines

GOLD guidelines

The 2018 clinical practice guidelines from the GOLD report on COPD are summarized.[105, 109]

Diagnosis and initial assessment recommendations are as follows:

Prevention and maintenance therapy recommendations are as follows:

Stable COPD recommendations are as follows:

Exacerbation recommendations are as follows:

COPD and comorbidity recommendations are as follows:

In the 2016 update of the GOLD guidelines, a rubric is used that assesses symptoms, breathlessness, spirometric classification, and risk of exacerbations to classify patients according to the following groups[4] :

The GOLD patient group-based management recommendations include the following[4] :

VA/DoD guidelines

In 2014, the VA/DoD released updated guidelines for the management of COPD.[106] These guidelines were endorsed with qualifications by the Institute of Clinical Systems Improvement (ICSI) in 2016.[110] The VA/DoD guidelines classify patients with COPD into the following two groups:

Major management recommendations include the following:

ACP, ACCP, ATS, and ERS joint guidelines

In 2011, the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society issued joint guidelines for the diagnosis and management of stable COPD. The major recommendations include the following[34] :

NIPPV Guidelines

In 2011, the Canadian Critical Care Trials Group and the Canadian Critical Care Society Noninvasive Ventilation Guidelines Group issued guidelines encouraging the use of noninvasive ventilation (NIPPV or continuous positive airway pressure [CPAP]) to avoid intubation for patients in acute care with respiratory failure. Key recommendations relevant to COPD include the following[107] :

Commentary accompanying the Canadian Critical Care guideline urges close patient monitoring and 24-hour availability of an experienced rescue team in case noninvasive ventilation fails and rapid intervention is required.[107]

Acute Exacerbation Guidelines

In 2015, the American College of Chest Physicians and Canadian Thoracic Society released guidelines on the prevention of acute exacerbations of COPD.[108]

Major recommendations include the following:

In patients with moderate-to-severe COPD, recommendations are as follows:

In patients with stable, moderate, severe, and very severe COPD, recommendations are as follows:

In patients with stable COPD, recommendations are as follows:

In patients aged 40 years who are smokers or have a history of smoking, recommendations are as follows:

Medication Summary

Oral and inhaled medications are used for patients with stable chronic obstructive pulmonary disease (COPD) to reduce dyspnea, improve exercise tolerance, and prevent complications. Most of the medications used in COPD treatment are directed at the following 4 potentially reversible mechanisms of airflow limitation:

Bronchodilators act to decrease muscle tone in small and large airways in the lungs, thereby increasing ventilation. The category includes beta2-adrenergic agonists, methylxanthines, and cholinergic/muscarinic antagonists.

Additionally, opioids have been shown in multiple studies to relieve dyspnea, particularly near the end of life. Dosage is very patient specific. Currow et al used a low, once-daily dose of sustained-release morphine for chronic refractory dyspnea.[111]

Use of mepolizumab has been associated with a lower rate of exacerbation in a subgroup of COPD patients with eosinophilia predominance.[112]

Albuterol (Proventil HFA, Ventolin HFA, ProAir HFA)

Clinical Context:  Albuterol is a short acting beta-agonist used for bronchospasm refractory to epinephrine. It relaxes bronchial smooth muscle by action on beta2-receptors with little effect on cardiac muscle contractility. Most patients (even those who have no measurable increase in expiratory flow) benefit from treatment. Inhaled beta agonists are prescribed initially as needed.

Metaproterenol

Clinical Context:  Metaproterenol relaxes bronchial smooth muscle by action on beta2-receptors with little effect on cardiac muscle contractility. Most patients (even those who have no measurable increase in expiratory flow) benefit from treatment. Inhaled beta agonists initially are prescribed as needed. Frequency may be increased.

Levalbuterol (Xopenex, Xopenex HFA)

Clinical Context:  Levalbuterol is a selective beta2-agonist agent used for the treatment or prevention of bronchospasm. Albuterol is a racemic mixture, while levalbuterol contains only the active R- enantiomer of albuterol. The S-enantiomer does not bind to beta2-receptors, but it may be responsible for some adverse effects of racemic albuterol, including bronchial hyperreactivity and reduced pulmonary function during prolonged use.

Class Summary

Beta2 -agonists activate specific B2 -adrenergic receptors on the surface of smooth muscle cells, which increases intracellular cyclic adenosine monophosphate (cAMP) and smooth muscle relaxation. Beta2 -agonists produce less bronchodilatation in COPD than in asthma. Patients use beta2 -agonists primarily for relief of symptoms of COPD. In patients with mild, intermittent symptoms, short-acting beta2 -agonists is recommended for symptomatic relief.

Salmeterol (Serevent Diskus)

Clinical Context:  By relaxing the smooth muscles of the bronchioles in conditions associated with bronchitis, emphysema, asthma, or bronchiectasis, salmeterol can relieve bronchospasms. The effect also may facilitate expectoration. It is shown to improve symptoms and morning peak flows. When administered at high or more frequent doses than recommended, incidence of adverse effects is higher. Bronchodilating effect lasts more than 12 hours. It is used on a fixed schedule in addition to regular use of anticholinergic agents.

Formoterol (Perforomist)

Clinical Context:  Formoterol relaxes the smooth muscles of the bronchioles and relieves bronchospasms. This effect also may facilitate expectoration. It is shown to improve symptoms and morning peak flows. When administered at high or more frequent doses than recommended, incidence of adverse effects is higher. Bronchodilating effect lasts more than 12 hours. It is used in addition to anticholinergic agents.

Arformoterol (Brovana)

Clinical Context:  Arformoterol is a selective, long-acting beta-2 adrenergic receptor agonist that has 2-fold greater potency than racemic formoterol. Pharmacologic effects of arformoterol are from the stimulation of intracellular adenyl cyclase, the enzyme that catalyzes the conversion of adenosine triphosphate to cyclic-3',5'-adenosine monophosphate (cAMP). Increases in intracellular cyclic AMP levels in turn cause relaxation of bronchial smooth muscles.

Indacaterol, inhaled (Arcapta Neohaler)

Clinical Context:  Long-acting beta2-agonist (LABA) indicated for long-term, once-daily maintenance bronchodilator treatment of airflow obstruction in patients with chronic obstructive pulmonary disease (COPD), including chronic bronchitis and/or emphysema. LABAs act locally in the lungs as bronchodilators. Stimulates intracellular adenyl cyclase, causing conversion of ATP to cyclic AMP; increased cyclic AMP levels cause relaxation of bronchial smooth muscle. Not for use as initial therapy in patients with acute deteriorating COPD.

Olodaterol inhaled (Striverdi Respimat)

Clinical Context:  Olodaterol is a once-daily LABA inhaler indicated for maintenance bronchodilator treatment in patients with COPD, including chronic bronchitis and/or emphysema in patients who are experiencing airflow obstruction. LABAs activate specific β2-adrenergic receptors on the surface of smooth muscle cells, which increases intracellular cAMP and smooth muscle relaxation.

Class Summary

Beta2 -agonist bronchodilators activate specific beta2 -adrenergic receptors on the surface of smooth muscle cells, which increases intracellular cyclic adenosine monophosphate (cAMP) and smooth muscle relaxation. In patients with more persistent symptoms, a long-acting beta agonist should be used. Long-acting beta agonists have been shown to increase exercise endurance, prevent nocturnal dyspnea, and improve quality of life. Long-acting beta-agonists include salmeterol, formoterol, arformoterol, and indacaterol. They all require twice-daily dosing, except for indacaterol and olodaterol, which are administered once daily.

Ipratropium (Atrovent HFA)

Clinical Context:  Short-acting anticholinergics, such as ipratropium bromide (Atrovent), have been shown to have equivalent or superior activity in patients with stable COPD when compared with a beta2 agonist. When combined with a beta2 agonist, a synergistic effect on bronchodilatation occurs. Typically, 2-4 puffs of ipratropium bromide are administered every 6-8 hours. This medication has a slower onset and a longer duration than a beta2 agonist and is less suitable for use on an as-needed basis.

Tiotropium (Spiriva)

Clinical Context:  Tiotropium, a bronchodilator similar to ipratropium, is a once-daily, long-acting anticholinergic medication. A quaternary ammonium compound, it elicits anticholinergic/antimuscarinic effects, with inhibitory effects on M3 receptors on airway smooth muscles, leading to bronchodilation.

Aclidinium (Tudorza Pressair)

Clinical Context:  Aclidinium is a twice-daily, long-acting selective muscarinic (M3) antagonist (anticholinergic) indicated for long-term maintenance of COPD including bronchitis and emphysema. It is available as breath-activated, dry powder metered-dose inhaler.

Umeclidinium bromide (Incruse Ellipta)

Clinical Context:  Umeclidinium bromide is a long-acting muscarinic antagonist (LAMA) inhalation powder, often referred to as an anticholinergic. It blocks action of acetylcholine at muscarinic receptors (M1 to M5) in the bronchial airways (M3) by preventing an increase in intracellular calcium concentration, leading to relaxation of airway smooth muscle, improved lung function, and decreased mucus secretion. Umeclidinium dissociates slowly from M3 muscarinic receptors extending its duration of action. It is indicated for the long-term, once-daily, maintenance treatment of airflow obstruction in patients with COPD), including chronic bronchitis and/or emphysema.

Revefenacin (Yupelri)

Clinical Context:  Revefenacin is a once-daily, long-acting muscarinic antagonist indicated for the maintenance treatment of COPD. It inhibits acetylcholine action at muscarinic receptors (M1 to M5), including muscarinic receptors on smooth muscle (M3) in the airway, leading to bronchodilation. It is administered by nebulizer using a mouthpiece.

Glycopyrrolate inhaled (Lonhala Magnair, Seebri Neohaler)

Clinical Context:  This agent contains glycopyrronium, which is a long-acting muscarinic antagonist (LAMA) that produces bronchodilation by inhibiting acetylcholine’s effect on the muscarinic receptor in the airway smooth muscle. It is indicated for long-term maintenance treatment of airflow obstruction in patients with COPD, including chronic bronchitis and/or emphysema. Seebri Neohaler is available as an encapsulated powder for inhalation that is used with the Neohaler device. Lonhala Magnair is available as a solution for nebulization used with the Magnair device.

Class Summary

Anticholinergic drugs compete with acetylcholine for postganglionic muscarinic receptors, thereby inhibiting cholinergically mediated bronchomotor tone, resulting in bronchodilatation. They block vagally mediated reflex arcs that cause bronchoconstriction. Clinical benefit is gained through a decrease in exercise-induced dynamic hyperinflation. These agents are poorly absorbed systemically and are relatively safe. Reported adverse effects include dry mouth, metallic taste, and prostatic symptoms.

Theophylline (Elixophyllin, Theo-24, Theochron)

Clinical Context:  Theophylline is a nonspecific phosphodiesterase inhibitor and is now limited to use as an adjunctive agent. Theophylline has a narrow therapeutic window with significant adverse effects, including anxiety, tremors, insomnia, nausea, cardiac arrhythmia (particularly multifocal atrial tachycardia), and seizures. It is reserved for patients with hard-to-control COPD or for individuals who are not able to use inhaled agents effectively. Theophylline is metabolized primarily via the hepatic cytochrome P450 system, a process affected by age, cardiac status, and liver abnormalities. Serum levels of theophylline need to be monitored because of the potential for toxicity. The previously recommended target range of 15-20 mg/dL has now been reduced to 8-13 mg/dL.

Aminophylline

Clinical Context:  Methylxanthine; directly relaxes smooth muscles of respiratory tract. It is reserved for patients with hard-to-control COPD or for individuals who are not able to use inhaled agents effectively.

Class Summary

Xanthine derivatives such as theophylline relax the smooth muscles of the bronchi and pulmonary blood vessels. Inhibition of phosphodiesterase by these agents causes an increase in cyclic adenosine monophosphate (cAMP), causing the relaxation of bronchial smooth muscles.

Roflumilast (Daliresp)

Clinical Context:  Roflumilast is a selective phosphodiesterase-4 (PDE-4) inhibitor. The specific mechanism of action is not well defined but is thought to be related to the effects of increased intracellular cyclic AMP in lung cells. It is indicated to decrease the frequency of exacerbations or the worsening of symptoms from severe COPD.

Class Summary

Selective phosphodiesterase-4 (PDE-4) inhibitors reduce exacerbations, improve dyspnea, and increase lung function in patients with severe COPD.

Fluticasone inhaled (Flovent)

Clinical Context:  Fluticasone inhibits bronchoconstriction producing direct smooth muscle relaxation. It may decrease the number and activity of inflammatory cells, in turn decreasing airway hyperresponsiveness. Effectiveness in COPD is not established. Inhaled corticosteroids have a lesser role in the management of chronic bronchitis. Several studies demonstrate no benefit, although approximately half of patients who respond to oral steroids may benefit from inhaled agents.

Budesonide inhaled (Pulmicort, Pulmicort Flexhaler)

Clinical Context:  Budesonide inhibits bronchoconstriction, producing direct smooth muscle relaxation. It may decrease the number and activity of inflammatory cells, in turn decreasing airway hyperresponsiveness. It has extremely potent vasoconstrictive and anti-inflammatory activity, and it alters the level of inflammation in airways by inhibiting multiple types of inflammatory cells and decreasing production of cytokines and other mediators. It also decreases inflammation by suppressing migration of polymorphonuclear leukocytes and reversing capillary permeability. Effectiveness is not established in COPD.

Class Summary

In acute exacerbations, steroids improve symptoms and lung function. Inhaled corticosteroids provide a more direct route of administration to the airways and, similar to other inhaled agents, are only minimally absorbed. Inhaled steroids have fewer adverse effects than do oral agents, although they improve expiratory flows less effectively, even at high doses. These agents may be beneficial in slowing the rate of progression in a subset of patients with COPD who have rapid decline.

Prednisone (Rayos)

Clinical Context:  Prednisone may decrease inflammation by reversing increased capillary permeability and suppressing PMN activity.

Methylprednisolone (Solu-Medrol, Medrol, A-Methapred)

Clinical Context:  Methylprednisolone is usually given in IV form for initiation of corticosteroid therapy, although the oral form is theoretically equally efficacious. The 2 forms are equal in potency, time of onset, and adverse effects. Inhaled corticosteroids are probably equally efficacious and have fewer adverse effects.

Class Summary

The use of oral steroids in the treatment of acute exacerbations is widely accepted and recommended, given their high efficacy. Note that oral steroids are not as effective in treating COPD exacerbations as they are in treating bronchial asthma exacerbations.

Albuterol/ipratropium (Combivent Respimat)

Clinical Context:  Ipratropium is chemically related to atropine, and it has antisecretory properties. Albuterol is a beta agonist for bronchospasm refractory to epinephrine. It relaxes bronchial smooth muscle by action on beta2 receptors with little effect on cardiac muscle contractility.

Metered-dose inhalers that contain chlorofluorocarbons (CFCs) are currently being phased out in the US; alternate inhalers without CFCs are available (eg, Combivent Respimat). This combination is also available as a nebulized solution.

Umeclidinium bromide/vilanterol inhaled (Anoro Ellipta)

Clinical Context:  Umeclidinium bromide and vilanterol is a long-acting muscarinic antagonist (LAMA) and long-acting beta2-agonist (LABA) inhalation powder. It is the first once-daily dual bronchodilator approved. It is indicated for long-term maintenance treatment of airflow obstruction in patients with COPD, including chronic bronchitis and/or emphysema.

Tiotropium/olodaterol inhaled (Stiolto Respimat)

Clinical Context:  Tiotropium/olodaterol inhaled (Stiolto Respimat) is a combination product containing olodaterol, a long-acting beta2-adrenergic agonist (LABA) plus tiotropium, a long-acting antimuscarinic agent. It is indicated for long-term, once-daily maintenance treatment of airflow obstruction in patients with COPD, including chronic bronchitis and/or emphysema.

Indacaterol, inhaled/glycopyrrolate inhaled (Utibron Neohaler)

Clinical Context:  Contains glycopyrronium, which is a long-acting muscarinic antagonist (LAMA) that produces bronchodilation by inhibiting acetylcholine’s effect on muscarinic receptor in the airway smooth muscle. Also contains indacaterol, a long-acting beta2-agonist (LABA) that stimulates intracellular adenyl cyclase, causing conversion of ATP to cyclic AMP, and thereby relaxes bronchial smooth muscle. It is indicated for long-term maintenance treatment of airflow obstruction in patients with COPD, including chronic bronchitis and/or emphysema.

Glycopyrrolate inhaled/formoterol (Bevespi Aerosphere)

Clinical Context:  This agent is a combination inhaler with glycopyrrolate, a long-acting muscarinic antagonist (LAMA), often referred to as an anticholinergic, and a long-acting beta2-agonist (LABA) with a rapid onset of action. Pharmacologic effect results in bronchodilation and relaxation of bronchial smooth muscle. It is indicated for the long-term, maintenance treatment of airflow obstruction with COPD, including chronic bronchitis and/or emphysema.

Aclidinium/formoterol (Duaklir)

Clinical Context:  Aclidinium is a long-acting muscarinic antagonist (LAMA), often referred to as an anticholinergic, with specificity for muscarinic receptors with affinity for the M3 (subscript) receptor in the airways. It produces bronchodilation by inhibiting acetylcholine’s effect on muscarinic receptors in the airway smooth muscle. 

Formoterol is a long-acting selective beta-2 agonist (LABA). It elicits bronchial smooth muscle relaxation by stimulation of intracellular adenyl cyclase, the enzyme that catalyzes the conversion of adenosine triphosphate (ATP) to cyclic-3', 5'-adenosine monophosphate (cyclic AMP). Increased cyclic AMP levels cause relaxation of bronchial smooth muscle and inhibit release of mediators of immediate hypersensitivity from cells, especially from mast cells.

It is indicated for maintenance treatment of COPD.

Class Summary

These agents combine the benefits of a beta-adrenergic agonist with the prolonged action of an anticholinergic agent.

Budesonide/formoterol (Symbicort)

Clinical Context:  Formoterol relieves bronchospasms by relaxing the smooth muscles of the bronchioles in conditions associated with asthma. Budesonide is an inhaled corticosteroid that alters the level of inflammation in airways by inhibiting multiple types of inflammatory cells and decreasing production of cytokines and other mediators involved in the asthmatic response.

Fluticasone and salmeterol (Advair Diskus)

Clinical Context:  Fluticasone inhibits bronchoconstriction mechanisms, producing direct smooth muscle relaxation. It may decrease number and activity of inflammatory cells, in turn decreasing airway hyperresponsiveness. It also has vasoconstrictive activity. Salmeterol relaxes the smooth muscles of the bronchioles in conditions associated with bronchitis, emphysema, asthma, or bronchiectasis and can relieve bronchospasms. Its effect may also facilitate expectoration.

Vilanterol/fluticasone inhaled (Breo Ellipta)

Clinical Context:  LABA and corticosteroid combination inhaler indicated for long-term, once-daily, maintenance treatment of airflow obstruction with COPD, including chronic bronchitis and/or emphysema. It is also approved to reduce COPD exacerbations. The product contains fluticasone fumarate, which has shown in vitro to exhibit a binding affinity for the human glucocorticoid receptor that is approximately 29.9 times that of dexamethasone and 1.7 times that of fluticasone propionate.

Class Summary

Combination therapy is recommended when COPD patients are uncontrolled with bronchodilator monotherapy. Agents that use a long-acting beta agonist and an inhaled corticosteroid are commonly used in asthma and COPD and show increased clinical benefits.

Amoxicillin (Moxatag)

Clinical Context:  Amoxicillin interferes with synthesis of cell wall mucopeptides during active multiplication, resulting in bactericidal activity against susceptible bacteria.

Doxycycline (Doryx, Monodox, Doxy 100, Adoxa)

Clinical Context:  Doxycycline is a broad-spectrum, synthetically derived bacteriostatic antibiotic in the tetracycline class. It is almost completely absorbed, it concentrates in bile, and it is excreted in urine and feces as a biologically active metabolite in high concentrations. It inhibits protein synthesis and, thus, bacterial growth by binding to 30S and possibly 50S ribosomal subunits of susceptible bacteria. It may block dissociation of peptidyl t-RNA from ribosomes, causing RNA-dependent protein synthesis to arrest.

Trimethoprim/sulfamethoxazole (Bactrim, Bactrim DS, Septra DS)

Clinical Context:  Sulfamethoxazole and trimethoprim inhibit bacterial synthesis of dihydrofolic acid by competing with para-aminobenzoic acid, resulting in inhibition of bacterial growth. Antibacterial activity of TMP-SMZ includes common urinary tract pathogens, except Pseudomonas aeruginosa. Like tetracycline, it has in vitro activity against Bartonella pertussis. It is not useful in mycoplasmal infections.

Cefuroxime (Zinacef, Ceftin)

Clinical Context:  Cefuroxime is a second-generation cephalosporin and maintains gram-positive activity of first-generation cephalosporins; it adds activity against P mirabilis, H influenzae, E coli, K pneumoniae, and M catarrhalis. It binds to penicillin-binding proteins and inhibits the final transpeptidation step of peptidoglycan synthesis, resulting in cell wall death. The condition of the patient, the severity of infection, and the susceptibility of the microorganism determine proper dose and route of administration. It also resists degradation by beta-lactamase.

Azithromycin (Zithromax, Zmax)

Clinical Context:  Azithromycin acts by binding to 50S ribosomal subunit of susceptible microorganisms and blocks dissociation of peptidyl t-RNA from ribosomes, causing RNA-dependent protein synthesis to arrest. It is used to treat acute bacterial exacerbations of chronic obstructive pulmonary disease caused by S pneumoniae, H influenzae, and M catarrhalis. They cover most potential etiologic agents, including Mycoplasma. The newer macrolides offer decreased GI upset as well as potential for improved compliance through reduced dosing frequency. They also afford improved action against H influenzae.

Clarithromycin (Biaxin)

Clinical Context:  Clarithromycin is a semisynthetic macrolide antibiotic that reversibly binds to the P site of the 50S ribosomal subunit of susceptible organisms and may inhibit RNA-dependent protein synthesis by stimulating dissociation of peptidyl t-RNA from ribosomes, causing bacterial growth inhibition.

Class Summary

In patients with COPD, chronic infection or colonization of the lower airways is common from S pneumoniae, H influenzae, and M catarrhalis. In patients with chronic severe airway obstruction, P aeruginosa infection may also be prevalent. The use of antibiotics for the treatment of acute exacerbations is well supported.

Nicotine transdermal system (Nicoderm CQ)

Clinical Context:  Transdermal nicotine patches are readily available for replacement therapy. Long-term success rates are 22-42%, compared with 2-25% for placebo. These agents are well tolerated, and the adverse effects are limited to local skin reactions. Nicotine replacement therapy patches are sold under the trade names NicoDerm, Nicotrol, and Habitrol. Each of these products is dosed with a scheduled graduated decrease in nicotine over 6-10 weeks.

Nicotine gum (Nicorette Gum)

Clinical Context:  Nicotine polacrilex is a chewing gum and has better quit rates than does counseling alone. Nicotine-replacement-therapy chewing pieces are marketed in 2 strengths (2 mg, 4 mg). An individual who smokes 1 pack per day should use 4-mg pieces. The 2-mg pieces are to be used by individuals who smoke less than 1 pack per day. Instruct patients to chew hourly and also to chew when needed for their initial cravings for 2 weeks. Gradually reduce the amount chewed over the next 3 months.

Bupropion (Zyban)

Clinical Context:  The use of the antidepressant bupropion (Zyban) is also effective for smoking cessation. This non-nicotine aid to smoking cessation enhances central nervous system nonadrenergic function. Bupropion may also be effective in patients who have not been able to quit smoking with nicotine replacement therapy. It is used in conjunction with a support group and/or behavioral counseling. It inhibits neuronal dopamine reuptake in addition to being a weak blocker of serotonin and norepinephrine reuptake.

Varenicline (Chantix)

Clinical Context:  Varenicline is a partial agonist selective for alpha4, beta2 nicotinic acetylcholine receptors. Its action is thought to result from activity at a nicotinic receptor subtype, where its binding produces agonist activity while simultaneously preventing nicotine binding. Agonistic activity is significantly lower than nicotine.

Class Summary

Smoking cessation continues to be the most important therapeutic intervention for COPD. Supervised use of pharmacologic agents is an important adjunct to self-help and group smoking cessation programs. Nicotine replacement therapies after smoking cessation reduce withdrawal symptoms.

Umeclidinium bromide/vilanterol inhaled/fluticasone furoate inhaled (Trelegy Ellipta)

Clinical Context:  The single-inhaler triple therapy is indicated for maintenance treatment of patients with COPD who are on a fixed-dose combination of fluticasone furoate and vilanterol for airflow obstruction and reducing exacerbations in whom additional treatment of airflow obstruction is desired, as well as for patients who are already receiving umeclidinium and a fixed-dose combination of furoate and vilanterol. The fixed-dose combination is available as furoate/umeclidinium/vilanterol 100/62.5/25 mcg.

Class Summary

COPD is generally treated with multiple inhaler therapies every day. Combination therapies improve drug adherence and provide significant therapeutic convenience. In September 2017, the FDA approved fluticasone furoate/umeclidinium/vilanterol, the first once-daily product approved in the United States that combines three active molecules into a single inhaler for COPD.

What is chronic obstructive pulmonary disease (COPD), and how common is it in the US?What are the signs and symptoms of chronic obstructive pulmonary disease (COPD)?What signs of chronic obstructive pulmonary disease (COPD) are observed in a thoracic exam?Which characteristics differentiate chronic bronchitis and emphysema in patients presenting with chronic obstructive pulmonary disease (COPD)?Which office test is used to diagnose chronic obstructive pulmonary disease (COPD), and what are the stages?How are arterial blood gas (ABG) findings used to determine acuteness and severity of chronic obstructive pulmonary disease (COPD)?Which type of chest radiographs are used in patients with emphysema associated with chronic obstructive pulmonary disease (COPD), and what are the findings?What are the advantages of high-resolution CT scanning in the diagnosis of chronic obstructive pulmonary disease (COPD)?In addition to spirometry and imaging, what additional tests that can be used in the diagnosis of chronic obstructive pulmonary disease (COPD)?What are the management strategies for each stage of chronic obstructive pulmonary disease (COPD)?Which drugs are used to treat chronic obstructive pulmonary disease (COPD)?What are the elements of a pulmonary rehabilitation program in chronic obstructive pulmonary disease (COPD) management?When is hospitalization indicated in patients with chronic obstructive pulmonary disease (COPD)?How common is chronic obstructive pulmonary disease (COPD) in the US, and which conditions are included in the classic triad of COPD?How has our understanding of chronic bronchitis and emphysema in chronic obstructive pulmonary disease (COPD) evolved?How do the GOLD guidelines define chronic obstructive pulmonary disease (COPD)?Which types of medications are used in the treatment of chronic obstructive pulmonary disease (COPD)?What is the pathophysiology of emphysema in chronic obstructive pulmonary disease (COPD)?What is the pathogenesis of chronic obstructive pulmonary disease (COPD)?What is the pathophysiology of chronic bronchitis in chronic obstructive pulmonary disease (COPD)?What are the roles of emphysematous destruction and small airway inflammation in chronic obstructive pulmonary disease (COPD) pathophysiology?What is the role of dynamic hyperinflation in chronic obstructive pulmonary disease (COPD) pathophysiology?What is the role of cigarette smoking in chronic obstructive pulmonary disease (COPD)?Which environmental factors contribute to the development of chronic obstructive pulmonary disease (COPD)?What is the role of airway hyperresponsiveness (Dutch hypothesis) in the development of chronic obstructive pulmonary disease (COPD)?What is alpha1-antitrypsin (AAT), and what is its role in the development of chronic obstructive pulmonary disease (COPD)?How does IV drug use contribute to the development of chronic obstructive pulmonary disease (COPD)?What is the role of immunodeficiency syndromes in the development of chronic obstructive pulmonary disease (COPD)?Which inflammatory processes are associated with chronic obstructive pulmonary disease (COPD)?Which connective tissue disorders are associated with the development of chronic obstructive pulmonary disease (COPD)?What is Salla disease, and how is it associated with the development of chronic obstructive pulmonary disease (COPD)?How prevalent are emphysema and chronic bronchitis in the US, and why is the prevalence of chronic obstructive pulmonary disease (COPD) likely underestimated?What is the worldwide prevalence of chronic obstructive pulmonary disease (COPD)?Is chronic obstructive pulmonary disease (COPD) more common in men or women?What is the prevalence of chronic obstructive pulmonary disease (COPD) in patients with a history of smoking?What is the mortality rate associated with chronic obstructive pulmonary disease (COPD)?What is the BODE index, and how is used to estimate the prognosis of chronic obstructive pulmonary disease (COPD)?Are questionnaires effective in assessing quality of life in patients with chronic obstructive pulmonary disease (COPD)?What are the risk factors for mortality in patients with chronic obstructive pulmonary disease (COPD)?What can patients do to actively manage their chronic obstructive pulmonary disease (COPD)?When do patients with chronic obstructive pulmonary disease (COPD) typically seek treatment, and which signs and symptoms are observed at presentation?According to the COPD guidelines, what 3 signs indicate airflow obstruction in patients presenting with chronic obstructive pulmonary disease (COPD)?How does chronic obstructive pulmonary disease (COPD) progress, and which comorbidities contribute to mortality?What is the role of chronic obstructive pulmonary disease (COPD) in mild cognitive impairment (MCI)?How can the clinical and historical findings be used to differentiate the types of chronic obstructive pulmonary disease (COPD)?Which respiratory findings in a patient with chronic obstructive pulmonary disease (COPD) are associated with severe disease?How do the characteristics of chronic bronchitis and emphysema differ in chronic obstructive pulmonary disease (COPD)?What is the GOLD criteria for staging chronic obstructive pulmonary disease (COPD), and what other staging systems are used in COPD?How is congestive heart failure (CHF) differentiated from chronic obstructive pulmonary disease (COPD)?How is bronchiectasis differentiated from chronic obstructive pulmonary disease (COPD)?How is bronchiolitis obliterans distinguished from chronic obstructive pulmonary disease (COPD)?How is adult-onset asthma distinguished from chronic obstructive pulmonary disease (COPD)?What are the differential diagnoses for Chronic Obstructive Pulmonary Disease (COPD)?What is the defining feature of chronic obstructive pulmonary disease (COPD)?How is the formal diagnosis of chronic obstructive pulmonary disease (COPD) made?Are blood-based biomarkers used in the diagnosis of chronic obstructive pulmonary disease (COPD)?Why is chronic obstructive pulmonary disease (COPD) underdiagnosed in its early stages?How is arterial blood gas (ABG) analysis used to determine the acuteness and severity of chronic obstructive pulmonary disease (COPD)?How is pH level used to assess respiratory compromise in chronic obstructive pulmonary disease (COPD)?Which electrolyte levels should be monitored in patients with chronic obstructive pulmonary disease (COPD)?What is the role of alpha1-antitrypsin (AAT) testing in the workup of chronic obstructive pulmonary disease (COPD)?What are the characteristics of sputum in chronic obstructive pulmonary disease (COPD)?How does B-type natriuretic peptide (BNP) bind?What is the role of B-type natriuretic peptide (BNP) levels in evaluating chronic obstructive pulmonary disease (COPD) exacerbations?What do chest radiographs demonstrate in patients with severe chronic obstructive pulmonary disease (COPD)?How is high-resolution CT (HRCT) scanning used in patients with chronic obstructive pulmonary disease (COPD)?What are the causes and effects of pulmonary hypertension in patients with chronic obstructive pulmonary disease (COPD)?What is the role of echocardiography in the diagnosis of chronic obstructive pulmonary disease (COPD)?What is the role of pulmonary function tests in the workup of chronic obstructive pulmonary disease (COPD)?How is the 6-minute walking distance (6MWD) test used in the evaluation of patients with chronic obstructive pulmonary disease (COPD)?What is the role of pulse oximetry in the workup of chronic obstructive pulmonary disease (COPD)?What is the role of ECG in the workup of chronic obstructive pulmonary disease (COPD)?When is right-sided heart catheterization indicated in patients with chronic obstructive pulmonary disease (COPD)?What hematocrit level in chronic hypoxemia indicates polycythemia?How effective is aclidinium in the treatment of chronic obstructive pulmonary disease (COPD)?How does umeclidinium bromide work in the treatment of chronic obstructive pulmonary disease (COPD), and is it safe and effective?What is the goal of treatment in chronic obstructive pulmonary disease (COPD), and what options are available?What is the most effective disease management strategy for chronic obstructive pulmonary disease (COPD)?When should a patient with chronic obstructive pulmonary disease (COPD) be admitted to an ICU?What types of medications are used in the treatment of chronic obstructive pulmonary disease (COPD)?What is the role of nutrition in chronic obstructive pulmonary disease (COPD)?What is the role of bronchodilators in chronic obstructive pulmonary disease (COPD)?Which medications are most effective as first-line therapy in the treatment of chronic obstructive pulmonary disease (COPD)?How do beta2-agonist bronchodilators work to treat chronic obstructive pulmonary disease (COPD), and what are the potential adverse effects?How do anticholinergic drugs work to treat chronic obstructive pulmonary disease (COPD), and what are the potential adverse effects?What adverse effects of anticholinergic drugs have been demonstrated in men with chronic obstructive pulmonary disease (COPD)?What is the preferred method of delivery of medications in the treatment of chronic obstructive pulmonary disease (COPD)?What are the risks associated with using newly prescribed long-acting beta-agonist and anticholinergic treatments for chronic obstructive pulmonary disease (COPD)?Is combination or monotherapy more effective in the treatment of chronic obstructive pulmonary disease (COPD)?How does glycopyrrolate work to treat chronic obstructive pulmonary disease (COPD)?How does combination glycopyrrolate/formoterol therapy work to treat chronic obstructive pulmonary disease (COPD)?What long-term maintenance options are available for chronic obstructive pulmonary disease (COPD)?What is the efficacy of tiotropium in the treatment of chronic obstructive pulmonary disease (COPD)?What is the efficacy of aclidinium in the treatment of chronic obstructive pulmonary disease (COPD)?What is the efficacy of umeclidinium bromide in the treatment of chronic obstructive pulmonary disease (COPD)?What is the efficacy of revefenacin in the treatment of chronic obstructive pulmonary disease (COPD)?How do phosphodiesterase inhibitors work in the treatment of chronic obstructive pulmonary disease (COPD)?How is theophylline used in the treatment of chronic obstructive pulmonary disease (COPD)?How is roflumilast used in the treatment of chronic obstructive pulmonary disease (COPD)?How do endogenous opioids help patients with chronic obstructive pulmonary disease (COPD)?What is the role of beta-blockers in the treatment of patients with cardiovascular disease and chronic obstructive pulmonary disease (COPD)?What is the role of a smoking cessation plan in the treatment of chronic obstructive pulmonary disease (COPD)?What are the stages of smoking cessation?What resources are available to help patients with chronic obstructive pulmonary disease (COPD) stop smoking?What are the symptoms of nicotine withdrawal, and how can they be prevented?What nicotine replacement therapies are available that can help patients with chronic obstructive pulmonary disease (COPD) stop smoking?How is inflammation in chronic obstructive pulmonary disease (COPD) treated?What is the role of inhaled corticosteroids in the treatment of inflammation in chronic obstructive pulmonary disease (COPD)?How is the efficacy of inhaled corticosteroids on inflammation in chronic obstructive pulmonary disease (COPD) measured?What is the role of IV steroids in the treatment of inflammation associated with chronic obstructive pulmonary disease (COPD)?What is the role of NSAIDs in the treatment of inflammation in chronic obstructive pulmonary disease (COPD)?What is the role of azithromycin in the treatment of chronic obstructive pulmonary disease (COPD)?How is infection treated in chronic obstructive pulmonary disease (COPD)?How are sputum viscosity and secretion clearance managed in chronic obstructive pulmonary disease (COPD)?How are proton pump inhibitors (PPIs) used to treat exacerbations and the common cold in patients with chronic obstructive pulmonary disease (COPD)?How is hypoxemia treated in chronic obstructive pulmonary disease (COPD)?What causes carbon dioxide retention, and how can it be avoided in patients with chronic obstructive pulmonary disease (COPD)?What are the physical hazards of oxygen therapy in chronic obstructive pulmonary disease (COPD), and how can they be avoided?How is oxygen delivered in stable hypoxemic patients?What is noninvasive positive-pressure ventilation (NIPPV), and how is it used in the treatment of chronic obstructive pulmonary disease (COPD)?Which vaccinations are indicated to reduce infection in patients with chronic obstructive pulmonary disease (COPD)?What are the treatment options for alpha1-antitrypsin (AAT) deficiency in chronic obstructive pulmonary disease (COPD)?When is hospitalization indicated for patients with chronic obstructive pulmonary disease (COPD)?How common are acute exacerbations of chronic obstructive pulmonary disease (AECOPD), and how are they treated?When is assisted ventilation indicated in the treatment of chronic obstructive pulmonary disease (COPD)?When is bullectomy indicated in the treatment of chronic obstructive pulmonary disease (COPD)?What is the role of lung volume reduction surgery in the treatment of chronic obstructive pulmonary disease (COPD)?What is the role of lung transplantation in the treatment of chronic obstructive pulmonary disease (COPD)?What follow-up is indicated after an acute exacerbation of chronic obstructive pulmonary disease (AECOPD)?What are the main goals of pulmonary rehabilitation in chronic obstructive pulmonary disease (COPD)?How is a successful pulmonary rehabilitation program implemented in chronic obstructive pulmonary disease (COPD)?What are the benefits of a pulmonary rehabilitation program for patients with chronic obstructive pulmonary disease (COPD)?What is the typical approach to a pulmonary rehabilitation program for patients with chronic obstructive pulmonary disease (COPD)?What is the role of patient education in a pulmonary rehabilitation program for chronic obstructive pulmonary disease (COPD)?What is the role of exercise in a pulmonary rehabilitation program for chronic obstructive pulmonary disease (COPD)?What is the role of breathing exercises in patients with chronic obstructive pulmonary disease (COPD)?When is it safe for patients with chronic obstructive pulmonary disease (COPD) to fly, and what accommodations can be made when necessary?What is the role PaO2 measurement in determining if oxygen therapy is indicated for sleep in patients with chronic obstructive pulmonary disease (COPD)?What is the role of an end-of-life care discussion in patients with chronic obstructive pulmonary disease (COPD)?What guidelines exist regarding chronic obstructive pulmonary disease (COPD)?What are the screening guidelines for chronic obstructive pulmonary disease (COPD)?What are the guidelines for preventing tobacco use in patients who smoke or who have chronic obstructive pulmonary disease (COPD)?What are the GOLD guidelines for the general management of chronic obstructive pulmonary disease (COPD)?What are the GOLD guidelines for the classification of chronic obstructive pulmonary disease (COPD)?What are the GOLD patient group-based management recommendations for the treatment of patients with chronic obstructive pulmonary disease (COPD)?What are the VA/DoD guidelines for the management of chronic obstructive pulmonary disease (COPD)?What are the ACP, ACCP, ATS, and ERS joint guidelines for the diagnosis and management of stable chronic obstructive pulmonary disease (COPD)?What are the guidelines for the use of noninvasive ventilation in patients with chronic obstructive pulmonary disease (COPD)?What are the guidelines for the prevention of acute exacerbations in chronic obstructive pulmonary disease (COPD)?What are the guidelines for the prevention of acute exacerbations in moderate-to-severe chronic obstructive pulmonary disease (COPD)?What are the guidelines for the prevention of acute exacerbations in patients with any stage of chronic obstructive pulmonary disease (COPD)?What are the guidelines for the prevention of acute exacerbations in stable chronic obstructive pulmonary disease (COPD)?What are the guidelines for the prevention of acute exacerbations of chronic obstructive pulmonary disease (COPD) in patients aged 40 years or older who smoke or have a history of smoking?Which types of medications are used for the treatment of chronic obstructive pulmonary disease (COPD)?Which medications in the drug class Beta2-Adrenergic Agonists, Short-Acting are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Beta2-Adrenergic Agonists, Long-Acting are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Anticholinergics, Respiratory are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Xanthine Derivative are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Phosphodiesterase-4 Inhibitors are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Corticosteroids, Inhalant are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Corticosteroids, Oral are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Beta-Adrenergic Agonist and Anticholinergic Agent Combinations are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Beta2-Adrenergic Agonist and Corticosteroid Combinations are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Antibiotics are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Smoking Cessation Therapies are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?Which medications in the drug class Other Combinations are used in the treatment of Chronic Obstructive Pulmonary Disease (COPD)?

Author

Zab Mosenifar, MD, FACP, FCCP, Geri and Richard Brawerman Chair in Pulmonary and Critical Care Medicine, Professor and Executive Vice Chairman, Department of Medicine, Medical Director, Women's Guild Lung Institute, Cedars Sinai Medical Center, University of California, Los Angeles, David Geffen School of Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

Annie Harrington, MD, Fellow in Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center

Disclosure: Nothing to disclose.

Nader Kamangar, MD, FACP, FCCP, FCCM, Professor of Clinical Medicine, University of California, Los Angeles, David Geffen School of Medicine; Chief, Division of Pulmonary and Critical Care Medicine, Vice-Chair, Department of Medicine, Olive View-UCLA Medical Center

Disclosure: Nothing to disclose.

Nidhi S Nikhanj, MD, Fellow, Department of Pulmonary and Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles

Disclosure: Nothing to disclose.

Specialty Editors

Mary L Windle, PharmD, Adjunct Associate Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Nothing to disclose.

Chief Editor

John J Oppenheimer, MD, Clinical Professor, Department of Medicine, Rutgers New Jersey Medical School; Director of Clinical Research, Pulmonary and Allergy Associates, PA

Disclosure: Received research grant from: quintiles, PRA, ICON, Novartis: Adjudication<br/>Received consulting fee from AZ for consulting; Received consulting fee from Glaxo, Myelin, Meda for consulting; Received grant/research funds from Glaxo for independent contractor; Received consulting fee from Merck for consulting; Received honoraria from Annals of Allergy Asthma Immunology for none; Partner received honoraria from ABAI for none. for: Atlantic Health System.

Acknowledgements

Ryland P Byrd Jr, MD Professor, Department of Internal Medicine, Division of Pulmonary Medicine and Critical Care Medicine, Program Director of Pulmonary Diseases and Critical Care Medicine Fellowship, East Tennessee State University, James H Quillen College of Medicine; Medical Director of Respiratory Therapy, James H Quillen Veterans Affairs Medical Center

Ryland P Byrd Jr, MD is a member of the following medical societies: American College of Chest Physicians and American Thoracic Society

Disclosure: Nothing to disclose.

Sat Sharma, MD, FRCPC Professor and Head, Division of Pulmonary Medicine, Department of Internal Medicine, University of Manitoba; Site Director, Respiratory Medicine, St Boniface General Hospital

Sat Sharma, MD, FRCPC is a member of the following medical societies: American Academy of Sleep Medicine, American College of Chest Physicians, American College of Physicians-American Society of Internal Medicine, American Thoracic Society, Canadian Medical Association, Royal College of Physicians and Surgeons of Canada, Royal Society of Medicine, Society of Critical Care Medicine, and World Medical Association

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

  1. Brooks M. FDA Clears Olodaterol (Striverdi Respimat) for COPD. Medscape Medical News. Available at http://www.medscape.com/viewarticle/829248. Accessed: August 4, 2014.
  2. Maclay JD, Rabinovich RA, MacNee W. Update in chronic obstructive pulmonary disease 2008. Am J Respir Crit Care Med. 2009 Apr 1. 179(7):533-41. [View Abstract]
  3. Casanova C, Cote C, Marin JM, Pinto-Plata V, de Torres JP, Aguirre-Jaime A, et al. Distance and oxygen desaturation during the 6-min walk test as predictors of long-term mortality in patients with COPD. Chest. 2008 Oct. 134(4):746-52. [View Abstract]
  4. [Guideline] Global strategy for diagnosis, management, and prevention of COPD: 2016. Global Initiative for Chronic Obstructive Lung Disease. Available at http://goldcopd.org/gold-reports/ . Accessed: May 7, 2016.
  5. Feghali-Bostwick CA, Gadgil AS, Otterbein LE, Pilewski JM, Stoner MW, Csizmadia E. Autoantibodies in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. Jan 15 2008. 177(2):156-63.
  6. Houben JM, Mercken EM, Ketelslegers HB, Bast A, Wouters EF, Hageman GJ. Telomere shortening in chronic obstructive pulmonary disease. Respir Med. 2009 Feb. 103(2):230-6. [View Abstract]
  7. Morissette MC, Vachon-Beaudoin G, Parent J, Chakir J, Milot J. Increased p53 level, Bax/Bcl-x(L) ratio, and TRAIL receptor expression in human emphysema. Am J Respir Crit Care Med. 2008 Aug 1. 178(3):240-7. [View Abstract]
  8. Hodge S, Hodge G, Jersmann H, Matthews G, Ahern J, Holmes M, et al. Azithromycin improves macrophage phagocytic function and expression of mannose receptor in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008 Jul 15. 178(2):139-48. [View Abstract]
  9. Casanova C, de Torres JP, Navarro J, Aguirre-Jaime A, Toledo P, Cordoba E, et al. Microalbuminuria and hypoxemia in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010 Oct 15. 182(8):1004-10. [View Abstract]
  10. Ofir D, Laveneziana P, Webb KA, Lam YM, O'Donnell DE. Mechanisms of dyspnea during cycle exercise in symptomatic patients with GOLD stage I chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2008 Mar 15. 177(6):622-9. [View Abstract]
  11. Belman MJ, Botnick WC, Shin JW. Inhaled bronchodilators reduce dynamic hyperinflation during exercise in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1996 Mar. 153(3):967-75. [View Abstract]
  12. O'Donnell DE, Lam M, Webb KA. Spirometric correlates of improvement in exercise performance after anticholinergic therapy in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1999 Aug. 160(2):542-9. [View Abstract]
  13. Marin JM, Carrizo SJ, Gascon M, Sanchez A, Gallego B, Celli BR. Inspiratory capacity, dynamic hyperinflation, breathlessness, and exercise performance during the 6-minute-walk test in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2001 May. 163(6):1395-9. [View Abstract]
  14. O'Donnell DE, Fluge T, Gerken F, Hamilton A, Webb K, Aguilaniu B. Effects of tiotropium on lung hyperinflation, dyspnoea and exercise tolerance in COPD. Eur Respir J. 2004 Jun. 23(6):832-40. [View Abstract]
  15. Martinez FJ, de Oca MM, Whyte RI, Stetz J, Gay SE, Celli BR. Lung-volume reduction improves dyspnea, dynamic hyperinflation, and respiratory muscle function. Am J Respir Crit Care Med. 1997 Jun. 155(6):1984-90. [View Abstract]
  16. Celli BR. Update on the management of COPD. Chest. 2008 Jun. 133(6):1451-62. [View Abstract]
  17. Casanova C, Cote C, de Torres JP, Aguirre-Jaime A, Marin JM, Pinto-Plata V. Inspiratory-to-total lung capacity ratio predicts mortality in patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2005 Mar 15. 171(6):591-7. [View Abstract]
  18. Nagelmann A, Tonnov A, Laks T, Sepper R, Prikk K. Lung Dysfunction of Chronic Smokers with No Signs of COPD. COPD. 2011 Apr 22. [View Abstract]
  19. Lamprecht B, McBurnie MA, Vollmer WM, Gudmundsson G, Welte T, Nizankowska-Mogilnicka E, et al. COPD in Never Smokers: Results From the Population-Based Burden of Obstructive Lung Disease Study. Chest. 2011 Apr. 139(4):752-763. [View Abstract]
  20. Andersen ZJ, Hvidberg M, Jensen SS, Ketzel M, Loft S, Sorensen M, et al. Chronic obstructive pulmonary disease and long-term exposure to traffic-related air pollution: a cohort study. Am J Respir Crit Care Med. 2011 Feb 15. 183(4):455-61. [View Abstract]
  21. Tashkin DP, Altose MD, Bleecker ER, Connett JE, Kanner RE, Lee WW, et al. The lung health study: airway responsiveness to inhaled methacholine in smokers with mild to moderate airflow limitation. The Lung Health Study Research Group. Am Rev Respir Dis. 1992 Feb. 145 (2 Pt 1):301-10. [View Abstract]
  22. Crothers K, Butt AA, Gibert CL, Rodriguez-Barradas MC, Crystal S, Justice AC. Increased COPD among HIV-positive compared to HIV-negative veterans. Chest. 2006 Nov. 130(5):1326-33. [View Abstract]
  23. Adams PF, Barnes PM, Vickerie JL. Summary health statistics for the U.S. population: National Health Interview Survey, 2007. Vital Health Stat 10. 2008 Nov. 1-104. [View Abstract]
  24. Buist AS, McBurnie MA, Vollmer WM, Gillespie S, Burney P, Mannino DM, et al. International variation in the prevalence of COPD (the BOLD Study): a population-based prevalence study. Lancet. 2007 Sep 1. 370(9589):741-50. [View Abstract]
  25. Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, Mannino DM. Global burden of COPD: systematic review and meta-analysis. Eur Respir J. 2006 Sep. 28(3):523-32. [View Abstract]
  26. Foreman MG, Zhang L, Murphy J, et al. Early-Onset COPD is Associated with Female Gender, Maternal Factors, and African American Race in the COPDGene Study. Am J Respir Crit Care Med. 2011 May 11. [View Abstract]
  27. Mintz ML, Yawn BP, Mannino DM, et al. Prevalence of airway obstruction assessed by lung function questionnaire. Mayo Clin Proc. 2011 May. 86(5):375-81. [View Abstract]
  28. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca M, Mendez RA, et al. The body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med. 2004 Mar 4. 350(10):1005-12. [View Abstract]
  29. Waschki B, Kirsten A, Holz O, et al. Physical activity is the strongest predictor of all-cause mortality in patients with COPD: a prospective cohort study. Chest. 2011 Aug. 140(2):331-42. [View Abstract]
  30. Abrams TE, Vaughan-Sarrazin M, Fan VS, Kaboli PJ. Geographic isolation and the risk for chronic obstructive pulmonary disease-related mortality: a cohort study. Ann Intern Med. 2011 Jul 19. 155(2):80-6. [View Abstract]
  31. Sundh J, Stallberg B, Lisspers K, Montgomery SM, Janson C. Co-Morbidity, Body Mass Index and Quality of Life in COPD Using the Clinical COPD Questionnaire. COPD. 2011 Apr 22. [View Abstract]
  32. McNamara D. Bronchiectasis Linked to Higher Mortality in COPD Patients. Medscape Medical News. February 8, 2013.
  33. Martinez-Garcia MA, de la Rosa D, Soler-Cataluna JJ, Donat-Sanz Y, Catalan Serra P, Agramunt Lerma M, et al. Prognostic Value of Bronchiectasis in Patients with Moderate-to-Severe Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2013 Feb 7. [View Abstract]
  34. [Guideline] Qaseem A, Wilt TJ, Weinberger SE, et al. Diagnosis and Management of Stable Chronic Obstructive Pulmonary Disease: A Clinical Practice Guideline Update from the American College of Physicians, American College of Chest Physicians, American Thoracic Society, and European Respiratory Society. Ann Intern Med. 2011 Aug 2. 155(3):179-191. [View Abstract]
  35. Hurst JR, Vestbo J, Anzueto A, Locantore N, Mullerova H, Tal-Singer R, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010 Sep 16. 363(12):1128-38. [View Abstract]
  36. Casanova C, Cote C, Marin JM, Pinto-Plata V, de Torres JP, Aguirre-Jaime A, et al. Distance and oxygen desaturation during the 6-min walk test as predictors of long-term mortality in patients with COPD. Chest. 2008 Oct. 134(4):746-52. [View Abstract]
  37. Spitzer C, Koch B, Grabe HJ, et al. Association of airflow limitation with trauma exposure and post-traumatic stress disorder. Eur Respir J. 2011 May. 37(5):1068-75. [View Abstract]
  38. Singh B, Parsaik AK, Mielke MM, et al. Chronic obstructive pulmonary disease and association with mild cognitive impairment: the Mayo Clinic study of aging. Mayo Clin Proc. 2013 Nov. 88(11):1222-30. [View Abstract]
  39. Barclay L. COPD linked to cognitive impairment and memory loss. Medscape Medical News. December 12, 2013.
  40. Bronchial hyperresponsiveness in cardiac failure. N Engl J Med. 1989 Dec 21. 321 (25):1756-8. [View Abstract]
  41. Prosen G, Klemen P, Strnad M, Grmec S. Combination of lung ultrasound (a comet-tail sign) and N-terminal pro-brain natriuretic peptide in differentiating acute heart failure from chronic obstructive pulmonary disease and asthma as cause of acute dyspnea in prehospital emergency setting. Crit Care. 2011. 15(2):R114. [View Abstract]
  42. Sin DD, Miller BE, Duvoix A, et al. Serum PARC/CCL-18 Concentrations and Health Outcomes in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2011 May 1. 183(9):1187-1192. [View Abstract]
  43. Osterweil N. COPD: Clinicians Miss Myriad Chances to Spot It Early. Medscape Medical News. Available at http://www.medscape.com/viewarticle/820597. Accessed: February 18, 2014.
  44. Miller MR, Quanjer PH, Swanney MP, Ruppel G, Enright PL. Interpreting lung function data using 80% predicted and fixed thresholds misclassifies more than 20% of patients. Chest. 2011 Jan. 139(1):52-9. [View Abstract]
  45. Rice KL, Dewan N, Bloomfield HE, Grill J, Schult TM, Nelson DB, et al. Disease management program for chronic obstructive pulmonary disease: a randomized controlled trial. Am J Respir Crit Care Med. 2010 Oct 1. 182(7):890-6. [View Abstract]
  46. Dewan NA, Rice KL, Caldwell M, Hilleman DE. Economic Evaluation of a Disease Management Program for Chronic Obstructive Pulmonary Disease. COPD. 2011 Apr 22. [View Abstract]
  47. Stephenson A, Seitz D, Bell CM, et al. Inhaled anticholinergic drug therapy and the risk of acute urinary retention in chronic obstructive pulmonary disease: a population-based study. Arch Intern Med. 2011 May 23. 171(10):914-20. [View Abstract]
  48. Gershon A, Croxford R, Calzavara A, To T, Stanbrook MB, Upshur R, et al. Cardiovascular Safety of Inhaled Long-Acting Bronchodilators in Individuals With Chronic Obstructive Pulmonary Disease. JAMA Intern Med. 2013 May 20. 1-9. [View Abstract]
  49. Wood S. Inhaled Long-Acting Bronchodilators in COPD Flagged Again for CV Hazard. Medscape Medical News. Available at http://www.medscape.com/viewarticle/804441. Accessed: June 4, 2013.
  50. Canavan N. Dual-Action Bronchodilator Eases COPD Exacerbations. Medscape [serial online]. Available at http://www.medscape.com/viewarticle/810739. Accessed: September 16, 2013.
  51. Decramer ML, Chapman KR, Dahl R, et al. Once-daily indacaterol versus tiotropium for patients with severe chronic obstructive pulmonary disease (INVIGORATE): a randomised, blinded, parallel-group study. The Lancet Respiratory Medicine. Available at http://www.thelancet.com/journals/lanres/article/PIIS2213-2600(13)70158-9/abstract. Accessed: October 7, 2013.
  52. Macfarlane L. Indacaterol and Tiotropium Similar in Effect and Safety. Medscape [serial online]. Available at http://www.medscape.com/viewarticle/811871. Accessed: October 7, 2013.
  53. Mahler DA, Kerwin E, Ayers T, FowlerTaylor A, Maitra S, Thach C, et al. FLIGHT1 and FLIGHT2: Efficacy and Safety of QVA149 (Indacaterol/Glycopyrrolate) versus Its Monocomponents and Placebo in Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2015 Nov 1. 192 (9):1068-79. [View Abstract]
  54. Rabe K, Martinez F, Rodriguez-Roisin R, Fabbri LM, Ferguson GT, Jones P, et al. PT003, a novel co-suspension MDI glycopyrronium/formoterol fixed-dose combination is superior to monocomponents in patients with COPD. Eur Respir J. 2015 Sep 01. 46(suppl 59):
  55. US Food and Drug Administration. FDA approves Anoro Ellipta to treat chronic obstructive pulmonary disease [press release]. December 18, 2013. Available at http://www.fda.gov/newsevents/newsroom/pressannouncements/ucm379057.htm. Accessed: December 30, 2013.
  56. Brown T. FDA approves umeclidinium and vilanterol combo for COPD. Medscape Medical News. December 18, 2013.
  57. Donohue JF, Maleki-Yazdi MR, Kilbride S, Mehta R, Kalberg C, Church A. Efficacy and safety of once-daily umeclidinium/vilanterol 62.5/25 mcg in COPD. Respir Med. 2013 Oct. 107(10):1538-46. [View Abstract]
  58. Koch A, Pizzichini E, Hamilton A, Hart L, Korducki L, De Salvo MC. Lung function efficacy and symptomatic benefit of olodaterol once daily delivered via Respimat versus placebo and formoterol twice daily in patients with GOLD 2-4 COPD: results from two replicate 48-week studies. Int J Chron Obstruct Pulmon Dis. 2014. 9:697-714. [View Abstract]
  59. Ferguson GT, Feldman GJ, Hofbauer P, Hamilton A, Allen L, Korducki L, et al. Efficacy and safety of olodaterol once daily delivered via Respimat in patients with GOLD 2-4 COPD: results from two replicate 48-week studies. Int J Chron Obstruct Pulmon Dis. 2014. 9:629-45. [View Abstract]
  60. Casaburi R, Mahler DA, Jones PW, Wanner A, San PG, ZuWallack RL, et al. A long-term evaluation of once-daily inhaled tiotropium in chronic obstructive pulmonary disease. Eur Respir J. 2002 Feb. 19(2):217-24. [View Abstract]
  61. Donohue JF, van Noord JA, Bateman ED, Langley SJ, Lee A, Witek TJ Jr, et al. A 6-month, placebo-controlled study comparing lung function and health status changes in COPD patients treated with tiotropium or salmeterol. Chest. 2002 Jul. 122(1):47-55. [View Abstract]
  62. Vincken W, van Noord JA, Greefhorst AP, Bantje TA, Kesten S, Korducki L, et al. Improved health outcomes in patients with COPD during 1 yr's treatment with tiotropium. Eur Respir J. 2002 Feb. 19(2):209-16. [View Abstract]
  63. Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med. 2008 Oct 9. 359(15):1543-54. [View Abstract]
  64. Brusasco V, Hodder R, Miravitlles M, Korducki L, Towse L, Kesten S. Health outcomes following treatment for six months with once daily tiotropium compared with twice daily salmeterol in patients with COPD. Thorax. 2003 May. 58(5):399-404. [View Abstract]
  65. Vogelmeier C, Hederer B, Glaab T, Schmidt H, Rutten-van Molken MP, Beeh KM, et al. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011 Mar 24. 364(12):1093-103. [View Abstract]
  66. Singh S, Loke YK, Enright PL, Furberg CD. Mortality associated with tiotropium mist inhaler in patients with chronic obstructive pulmonary disease: systematic review and meta-analysis of randomised controlled trials. BMJ. 2011 Jun 14. 342:d3215. [View Abstract]
  67. Jones PW, Rennard SI, Agusti A, Chanez P, Magnussen H, Fabbri L, et al. Efficacy and safety of once-daily aclidinium in chronic obstructive pulmonary disease. Respir Res. 2011 Apr 26. 12:55. [View Abstract]
  68. Hand L. FDA OKs Umeclidinium (Incruse Ellipta) for COPD. Medscape [serial online]. Available at http://www.medscape.com/viewarticle/824419. Accessed: May 4, 2014.
  69. Anoro Ellipta (umeclidinium and vilanterol inhalation powder) [package insert]. Research Triangle Park, NC: GlaxoSmithKline. 2013. Available at
  70. Kerwin EM, Donohue JF, Sethi S, Haumann B, Pendyala S, Dean L, et al. Revefenacin, a once-daily, long-acting muscarinic antagonist for nebulized therapy of chronic obstructive pulmonary disease (COPD): Results of a 52-week safety and tolerability phase 3 trial in participants with moderate to very severe COPD (poster A4239). Presented at the American Thoracic Society 2018 International Conference. San Diego, CA. May 21, 2018.
  71. Calverley PM, Rabe KF, Goehring UM, Kristiansen S, Fabbri LM, Martinez FJ. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet. 2009 Aug 29. 374(9691):685-94. [View Abstract]
  72. Gifford AH, Mahler DA, Waterman LA, et al. Neuromodulatory Effect of Endogenous Opioids on the Intensity and Unpleasantness of Breathlessness during Resistive Load Breathing in COPD. COPD. 2011 Apr 22. [View Abstract]
  73. Short PM, Lipworth SI, Elder DH, Schembri S, Lipworth BJ. Effect of beta blockers in treatment of chronic obstructive pulmonary disease: a retrospective cohort study. BMJ. 2011 May 10. 342:d2549. [View Abstract]
  74. Mottillo S, Filion KB, Belisle P, Joseph L, Gervais A, O'Loughlin J. Behavioural interventions for smoking cessation: a meta-analysis of randomized controlled trials. Eur Heart J. 2009 Mar. 30(6):718-30. [View Abstract]
  75. Wood-Baker RR, Gibson PG, Hannay M, Walters EH, Walters JA. Systemic corticosteroids for acute exacerbations of chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005. (1):CD001288. [View Abstract]
  76. Walters JA, Walters EH, Wood-Baker R. Oral corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005 Jul 20. CD005374. [View Abstract]
  77. Spencer S, Calverley PM, Burge PS, Jones PW. Impact of preventing exacerbations on deterioration of health status in COPD. Eur Respir J. 2004 May. 23(5):698-702. [View Abstract]
  78. Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007 Feb 22. 356(8):775-89. [View Abstract]
  79. Sin DD, Tashkin D, Zhang X, Radner F, Sjobring U, Thoren A. Budesonide and the risk of pneumonia: a meta-analysis of individual patient data. Lancet. 2009 Aug 29. 374(9691):712-9. [View Abstract]
  80. Pascoe S, Locantore N, Dransfield MT, Barnes NC, Pavord ID. Blood eosinophil counts, exacerbations, and response to the addition of inhaled fluticasone furoate to vilanterol in patients with chronic obstructive pulmonary disease: a secondary analysis of data from two parallel randomised controlled trials. Lancet Respir Med. 2015 Jun. 3 (6):435-42. [View Abstract]
  81. Siddiqui SH, Guasconi A, Vestbo J, Jones P, Agusti A, Paggiaro P, et al. Blood Eosinophils: A Biomarker of Response to Extrafine Beclomethasone/Formoterol in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med. 2015 Aug 15. 192 (4):523-5. [View Abstract]
  82. Bafadhel M, Peterson S, De Blas MA, Calverley PM, Rennard SI, Richter K, et al. Predictors of exacerbation risk and response to budesonide in patients with chronic obstructive pulmonary disease: a post-hoc analysis of three randomised trials. Lancet Respir Med. 2018 Feb. 6 (2):117-126. [View Abstract]
  83. Chen D, Restrepo MI, Fine MJ, et al. Observational study of inhaled corticosteroids on outcomes for COPD patients with pneumonia. Am J Respir Crit Care Med. 2011 Aug 1. 184(3):312-6. [View Abstract]
  84. Seemungal TA, Wilkinson TM, Hurst JR, Perera WR, Sapsford RJ, Wedzicha JA. Long-term erythromycin therapy is associated with decreased chronic obstructive pulmonary disease exacerbations. Am J Respir Crit Care Med. 2008 Dec 1. 178(11):1139-47. [View Abstract]
  85. Albert RK, Connett J, Bailey WC, et al. Azithromycin for prevention of exacerbations of COPD. N Engl J Med. 2011 Aug 25. 365(8):689-98. [View Abstract]
  86. Daniels JM, Snijders D, de Graaff CS, Vlaspolder F, Jansen HM, Boersma WG. Antibiotics in addition to systemic corticosteroids for acute exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010 Jan 15. 181(2):150-7. [View Abstract]
  87. Sasaki T, Nakayama K, Yasuda H, Yoshida M, Asamura T, Ohrui T, et al. A randomized, single-blind study of lansoprazole for the prevention of exacerbations of chronic obstructive pulmonary disease in older patients. J Am Geriatr Soc. 2009 Aug. 57(8):1453-7. [View Abstract]
  88. Duiverman ML, Wempe JB, Bladder G, Jansen DF, Kerstjens HA, Zijlstra JG. Nocturnal non-invasive ventilation in addition to rehabilitation in hypercapnic patients with COPD. Thorax. 2008 Dec. 63(12):1052-7. [View Abstract]
  89. Crockett AJ, Moss JR, Cranston JM, Alpers JH. Domicilary oxygen for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2000. (2):CD001744. [View Abstract]
  90. Ringbaek TJ. Continuous oxygen therapy for hypoxic pulmonary disease: guidelines, compliance and effects. Treat Respir Med. 2005. 4(6):397-408. [View Abstract]
  91. Sandland CJ, Morgan MD, Singh SJ. Patterns of domestic activity and ambulatory oxygen usage in COPD. Chest. 2008 Oct. 134(4):753-60. [View Abstract]
  92. Lightowler JV, Wedzicha JA, Elliott MW, Ram FS. Non-invasive positive pressure ventilation to treat respiratory failure resulting from exacerbations of chronic obstructive pulmonary disease: Cochrane systematic review and meta-analysis. BMJ. 2003 Jan 25. 326(7382):185. [View Abstract]
  93. Carrera M, Marin JM, Anton A, Chiner E, Alonso ML, Masa JF, et al. A controlled trial of noninvasive ventilation for chronic obstructive pulmonary disease exacerbations. J Crit Care. 2009 Sep. 24(3):473.e7-14. [View Abstract]
  94. Keenan SP, Kernerman PD, Cook DJ, Martin CM, McCormack D, Sibbald WJ. Effect of noninvasive positive pressure ventilation on mortality in patients admitted with acute respiratory failure: a meta-analysis. Crit Care Med. 1997 Oct. 25(10):1685-92. [View Abstract]
  95. Confalonieri M, Garuti G, Cattaruzza MS, Osborn JF, Antonelli M, Conti G. A chart of failure risk for noninvasive ventilation in patients with COPD exacerbation. Eur Respir J. 2005 Feb. 25(2):348-55. [View Abstract]
  96. Hubbard RC, Crystal RG. Augmentation therapy of alpha 1-antitrypsin deficiency. Eur Respir J Suppl. 1990 Mar. 9:44s-52s. [View Abstract]
  97. Hurst JR, Donaldson GC, Quint JK, Goldring JJ, Baghai-Ravary R, Wedzicha JA. Temporal clustering of exacerbations in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009 Mar 1. 179(5):369-74. [View Abstract]
  98. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, Ries A, et al. A randomized trial comparing lung-volume-reduction surgery with medical therapy for severe emphysema. N Engl J Med. 2003 May 22. 348(21):2059-73. [View Abstract]
  99. Titman A, Rogers CA, Bonser RS, Banner NR, Sharples LD. Disease-specific survival benefit of lung transplantation in adults: a national cohort study. Am J Transplant. 2009 Jul. 9(7):1640-9. [View Abstract]
  100. Burton CM, Milman N, Carlsen J, Arendrup H, Eliasen K, Andersen CB, et al. The Copenhagen National Lung Transplant Group: survival after single lung, double lung, and heart-lung transplantation. J Heart Lung Transplant. 2005 Nov. 24(11):1834-43. [View Abstract]
  101. Cote CG, Celli BR. Pulmonary rehabilitation and the BODE index in COPD. Eur Respir J. 2005 Oct. 26(4):630-6. [View Abstract]
  102. Dodd JW, Hogg L, Nolan J, et al. The COPD assessment test (CAT): response to pulmonary rehabilitation. A multicentre, prospective study. Thorax. 2011 May. 66(5):425-9. [View Abstract]
  103. [Guideline] Guirguis-Blake JM, Senger CA, Webber EM, Mularski RA, Whitlock EP. Screening for Chronic Obstructive Pulmonary Disease: Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA. 2016 Apr 5. 315 (13):1378-93. [View Abstract]
  104. [Guideline] US Preventive Services Task Force. Counseling and interventions to prevent tobacco use and tobacco-caused disease in adults and pregnant women: US Preventive Services Task Force Reaffirmation Recommendation Statement. Ann Int Med. Apr 21 2009. 150(8):551-555.
  105. [Guideline] Global Initiative for Chronic Obstructive Lung Disease. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Pulmonary Disease 2018 Report. Goldcopd.org. Available at https://goldcopd.org/wp-content/uploads/2017/11/GOLD-2018-v6.0-FINAL-revised-20-Nov_WMS.pdf. 2018; Accessed: October 23, 2018.
  106. [Guideline] Management of Chronic Obstructive Pulmonary Disease Working Group. VA/DoD clinical practice guideline for the management of chronic obstructive pulmonary disease. Washington (DC): Department of Veterans Affairs, Department of Defense. Available at http://www.healthquality.va.gov/guidelines/cd/copd/. December 2014; Accessed: May 7, 2016.
  107. [Guideline] Keenan SP, Sinuff T, Burns KE, Muscedere J, Kutsogiannis J, Mehta S, et al. Clinical practice guidelines for the use of noninvasive positive-pressure ventilation and noninvasive continuous positive airway pressure in the acute care setting. CMAJ. 2011 Feb 22. 183(3):E195-214. [View Abstract]
  108. [Guideline] Criner GJ, Bourbeau J, Diekemper RL, et al. Prevention of acute exacerbations of COPD: American College of Chest Physicians and Canadian Thoracic Society Guideline. Chest. 2015 Apr. 147 (4):894-942. [View Abstract]
  109. Mirza S, Clay RD, Koslow MA, Scanlon PD. COPD Guidelines: A Review of the 2018 GOLD Report. Mayo Clin Proc. 2018 Oct. 93 (10):1488-1502. [View Abstract]
  110. [Guideline] Institute for Clinical Systems Improvement. Diagnosis and Management of Chronic Obstructive Pulmonary Disease (COPD). 10th Edition. January 2016. Available at https://www.icsi.org/guidelines__more/catalog_guidelines_and_more/catalog_guidelines/catalog_respiratory_guidelines/copd/ . Accessed: May 6, 2016.
  111. Currow DC, McDonald C, Oaten S, Kenny B, Allcroft P, Frith P, et al. Once-daily opioids for chronic dyspnea: a dose increment and pharmacovigilance study. J Pain Symptom Manage. 2011 Sep. 42(3):388-99. [View Abstract]
  112. Pavord ID, Chanez P, Criner GJ, Kerstjens HAM, Korn S, Lugogo N, et al. Mepolizumab for Eosinophilic Chronic Obstructive Pulmonary Disease. N Engl J Med. 2017 Oct 26. 377 (17):1613-1629. [View Abstract]

Venn diagram of chronic obstructive pulmonary disease (COPD). Chronic obstructive lung disease is a disorder in which subsets of patients may have dominant features of chronic bronchitis, emphysema, or asthma. The result is airflow obstruction that is not fully reversible.

Venn diagram of chronic obstructive pulmonary disease (COPD). Chronic obstructive lung disease is a disorder in which subsets of patients may have dominant features of chronic bronchitis, emphysema, or asthma. The result is airflow obstruction that is not fully reversible.

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells.

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells (high-powered view).

Gross pathology of advanced emphysema. Large bullae are present on the surface of the lung.

Gross pathology of a patient with emphysema showing bullae on the surface.

At high magnification, loss of alveolar walls and dilatation of airspaces in emphysema can be seen.

Posteroanterior (PA) and lateral chest radiograph in a patient with severe chronic obstructive pulmonary disease (COPD). Hyperinflation, depressed diaphragm, increased retrosternal space, and hypovascularity of lung parenchyma are demonstrated.

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on lateral chest radiograph.

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on posteroanterior chest radiograph.

Severe bullous disease as seen on a computed tomography (CT) scan in a patient with chronic obstructive pulmonary disease (COPD).

Pressure volume curve comparing lungs with emphysema, lungs with restrictive disease, and normal lungs.

Flow volume curve of a patient with emphysema shows marked decrease in expiratory flow, hyperinflation, and air trapping (patient B) compared with a patient with restrictive lung disease, who has reduced lung volumes and preserved flow (patient A).

Forced expiratory volume in 1 second (FEV1) can be used to evaluate the prognosis in patients with emphysema. The benefit of smoking cessation is shown here because the deterioration in lung function parallels that of a nonsmoker, even in late stages of the disease. Redrawn from Fletcher C, Peato R. The natural history of chronic airflow obstruction. Br Med J 1977; 1: 1645-1648.

Oxygen therapy via nasal cannula.

Home supplemental oxygen.

Bilevel positive airway pressure (BiPAP).

Pulmonary rehabilitation.

Venn diagram of chronic obstructive pulmonary disease (COPD). Chronic obstructive lung disease is a disorder in which subsets of patients may have dominant features of chronic bronchitis, emphysema, or asthma. The result is airflow obstruction that is not fully reversible.

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells.

Histopathology of chronic bronchitis showing hyperplasia of mucous glands and infiltration of the airway wall with inflammatory cells (high-powered view).

Gross pathology of advanced emphysema. Large bullae are present on the surface of the lung.

Gross pathology of a patient with emphysema showing bullae on the surface.

At high magnification, loss of alveolar walls and dilatation of airspaces in emphysema can be seen.

Posteroanterior (PA) and lateral chest radiograph in a patient with severe chronic obstructive pulmonary disease (COPD). Hyperinflation, depressed diaphragm, increased retrosternal space, and hypovascularity of lung parenchyma are demonstrated.

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on lateral chest radiograph.

A lung with emphysema shows increased anteroposterior (AP) diameter, increased retrosternal airspace, and flattened diaphragm on posteroanterior chest radiograph.

Severe bullous disease as seen on a computed tomography (CT) scan in a patient with chronic obstructive pulmonary disease (COPD).

Pressure volume curve comparing lungs with emphysema, lungs with restrictive disease, and normal lungs.

Flow volume curve of a patient with emphysema shows marked decrease in expiratory flow, hyperinflation, and air trapping (patient B) compared with a patient with restrictive lung disease, who has reduced lung volumes and preserved flow (patient A).

Forced expiratory volume in 1 second (FEV1) can be used to evaluate the prognosis in patients with emphysema. The benefit of smoking cessation is shown here because the deterioration in lung function parallels that of a nonsmoker, even in late stages of the disease. Redrawn from Fletcher C, Peato R. The natural history of chronic airflow obstruction. Br Med J 1977; 1: 1645-1648.

Oxygen therapy via nasal cannula.

Home supplemental oxygen.

Bilevel positive airway pressure (BiPAP).

Pulmonary rehabilitation.

Chronic obstructive pulmonary disease (COPD). Pulmonary rehabilitation.

Chest radiograph of an emphysematous patient shows hyperinflated lungs with reduced vascular markings. Pulmonary hila are prominent, suggesting some degree of pulmonary hypertension (Correa da Silva, 2001).

Schematic representation of another sign of emphysema on the lateral chest radiograph. When the retrosternal space (defined as the space between the posterior border of the sternum and the anterior wall of the mediastinum) is larger than 2.5 cm, it is highly suggestive of overinflated lungs. This radiograph is from a patient with pectus carinatum, an important differential diagnosis to consider when this space is measured (Correa da Silva, 2001).

Close-up image shows emphysematous bullae in the left upper lobe. Note the subpleural, thin-walled, cystlike appearance (Correa da Silva, 2001).

A, Frontal posteroanterior (PA) chest radiograph shows no abnormality of the pulmonary vasculature, with normal intercostal spaces and a diaphragmatic dome between the 6th and 7th anterior ribs on both sides. B, Image in a patient with emphysema demonstrating reduced pulmonary vasculature resulting in hyperlucent lungs. The intercostal spaces are mildly enlarged, and the diaphragmatic domes are straightened and below the extremity of the seventh rib (Correa da Silva, 2001).

A, Lateral radiograph of the chest shows normal pulmonary vasculature, a retrosternal space within normal limits (&lt; 2.5 cm), and a normal angle between the diaphragm and the anterior thoracic wall. B, Lateral view of the chest shows increased pulmonary transparency, increased retrosternal space (>2.5 cm), and an angle between the thoracic wall and the diaphragm >90 degrees. Straightening of the diaphragm can be more evident in this projection than on others (Correa da Silva, 2001).

High-resolution CT (HRCT) in a patient after viral bronchiolitis obliterans demonstrates areas of airtrapping, which is predominant in the inferior lobes and associated with bronchiectasis in the left lower lobe. Note that the decreased attenuation caused by the airtrapping can simulate emphysema (Correa da Silva, 2001).

Pediatric high-resolution CT (HRCT) shows a hyperinflated right lung with large pulmonary bullae due to congenital lobar emphysema (Correa da Silva, 2001).

High-resolution CT (HRCT) demonstrates areas of centriacinar emphysema. Note the low attenuation areas without walls due to destruction of the alveoli septae centrally in the acini. Red element shows the size of a normal acinus (Correa da Silva, 2001).

High-resolution CT (HRCT) shows large bullae in both inferior lobes due to uniform enlargement and destruction of the alveoli walls causing distortion of the pulmonary architecture (Correa da Silva, 2001).

Panacinar emphysema of the left lung in a patient with a right lung transplant. Note the red element showing the size of a normal acinus and its discrepancy with the destroyed and enlarged airspaces of the left lower lobe (Correa da Silva, 2001).

High-resolution CT (HRCT) shows subpleural bullae consistent with paraseptal emphysema. Red mark shows the size of a normal acinus (Correa da Silva, 2001).

High-resolution CT (HRCT) shows enlarged air-spaces or bullae adjoining pulmonary scars, consistent with paracicatricial emphysema. Red mark shows the size of a normal acinus (Correa da Silva, 2001).

CT densitovolumetry of a nonsmoker, healthy young patient shows normal lungs. Less than 0.35% of lungs have attenuations below -950 HU (Correa da Silva, 2001).

Expiratory CT densitovolumetry shows no areas of airtrapping (Correa da Silva, 2001).

CT densitovolumetry in a patient with lung cancer. Three-dimensional (3D) image shows that the cancer is in the portion of the right lung that was less affected by emphysema in a patient with poor pulmonary function (Correa da Silva, 2001).

CT densitovolumetry shows the attenuation mask. Green areas are those with attenuation below the selected threshold (here, -950 HU to evaluate emphysema), and pink areas are those with attenuations above the threshold. Area outside the patient is highlighted in green because of air (Correa da Silva, 2001).

CT densitovolumetry demonstrates irregular distribution of the emphysema, with substantial predominance in the left lung (Correa da Silva, 2001).