ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Chronic obstructive pulmonary disease: Risk factors and risk reduction

Chronic obstructive pulmonary disease: Risk factors and risk reduction
Literature review current through: Jan 2024.
This topic last updated: Jan 04, 2024.

INTRODUCTION — Chronic obstructive pulmonary disease (COPD) is the result of complex interplay between clinical and molecular (ie, genetic) risk factors [1,2]. These interactions are the reason that two individuals may have identical clinical risk factors, but only one will develop COPD. Identifying risk factors for COPD and better understanding their interactions may lead to strategies that reduce the prevalence of COPD.

Risk factors for COPD and strategies for risk reduction are discussed in this topic review. The clinical manifestations, diagnosis, staging, natural history, and treatment of COPD are reviewed separately. (See "Chronic obstructive pulmonary disease: Diagnosis and staging" and "Chronic obstructive pulmonary disease: Prognostic factors and comorbid conditions" and "Stable COPD: Initial pharmacologic management".)

LUNG FUNCTION — Lung function normally increases as the lungs grow during childhood and adolescence, peaks shortly after 20 years of age, and then begins to decline gradually (figure 1). Risk factors for chronic obstructive pulmonary disease (COPD) cause one or more abnormal patterns of lung function as shown in the figure (figure 1):

Reduced lung growth

Premature lung function decline

Accelerated lung function decline

Accelerated lung function decline is most common. However, each of these abnormal patterns can reduce lung function to an extent that the individual is considered to have COPD. (See "Chronic obstructive pulmonary disease: Diagnosis and staging".)

CLINICAL RISK FACTORS — Definite risk factors for chronic obstructive pulmonary disease (COPD) include smoking and increased airway responsiveness [3-5]. Environmental exposures (other than smoking), atopy, antioxidant deficiency, and prior tuberculosis may also be risk factors [6]. A taxonomy of COPD types has been proposed based on these risk factors ("etiotypes") (table 1) [1,7].

Environmental exposures

Cigarette smoking — Numerous epidemiologic studies indicate that tobacco smoking is overwhelmingly the most important risk factor for COPD [6,8-14]. As an example, a retrospective cohort study (n = 8045) found that subjects who smoked cigarettes throughout a 25-year observation period were more likely than never smokers to develop COPD (36 versus 8 percent) [11]. In another study, the single best variable for predicting which adults will have airflow obstruction on spirometry is a history of more than 40 pack-years of smoking (positive likelihood ratio [LR], 12 [95% CI, 2.7-50]) [15,16]. However, other data suggest smoking duration may provide stronger risk estimates of COPD than the composite index of pack-years [17]. This may in part be due to increased risk associated with smoking exposure in early adolescence [18].

The presence of additional respiratory symptoms does not appear to increase risk of developing airflow obstruction in current or former smokers with significant tobacco exposure. In one longitudinal cohort of 495 patients aged 40 to 80 years with a more than 20 pack-year smoking history (median 40 pack-years) and preserved spirometry, risk of developing airway obstruction over five years was 30 to 35 percent regardless of the presence of respiratory symptoms [19].

Smoking tobacco through a Chinese water pipe (narghile) is also associated with an increased risk of COPD compared with never smoking (adjusted odds ratio [OR], 10.61; 95% CI, 6.89-16.34), challenging the assumption that filtering the tobacco smoke through water might be protective [20]. Smoking both tobacco and marijuana synergistically increases the risk of COPD and respiratory symptoms [21].

Genetic influences may enhance an individual's susceptibility to the detrimental effects of cigarette smoke. This is supported by an observational study that found that bronchodilator responsiveness (a surrogate measure of risk for accelerated lung function decline) was increased among current or former smokers who had a first-degree relative with severe early-onset COPD, compared to current or former smokers who did not have such a relative [22].

Pollution, biomass, and occupational exposures — Numerous studies indicate that environmental exposure to particulate matter, dusts, vapors, fumes, or organic antigens may also be a risk factor for COPD [6,13,23-38]. As an example, a population-based sample (n = 8515) found that COPD was more common among those exposed to occupational dust than those who were unexposed (odds ratio [OR] 1.5, 95% CI 1.17-2.08) [23]. Another study found that former (but not current) smokers who live in homes estimated to have high levels of indoor small (<2.5 micron) airborne particulate matter (PM2.5) demonstrate accelerated loss of lung function [39]. This finding may in part explain the observation that COPD is more common in women exposed to indoor biomass smoke [29,40,41].

In studies of outdoor environmental pollution, PM2.5 has been most strongly associated with incident COPD [42]. In one meta-analysis of worldwide observational studies using several different pollution exposure estimates, every 10 mcg/m3 increase in PM2.5 level was associated with an 18 percent increase in the incidence of COPD (pooled hazard ratio 1.18, 95% CI 1.13-1.23) [43]. Coarse particulates have often not been found to be strongly associated with COPD in observational studies [43,44]; variable results have been seen for nitrogen dioxide, ozone, and other components of air pollution [43,45,46].

Occupational exposure to chemical disinfectants (eg, glutaraldehyde, bleach, hydrogen peroxide, alcohol, and quaternary ammonium compounds) is associated with an increased risk of COPD among nurses; adjusted hazard ratios for high-level exposure range from 1.25 (95% CI 1.04-1.51) to 1.36 (95% CI 1.13-1.64) [47]. In a large biobank study, exposure to pesticides was also associated with COPD, with a prevalence ratio of 1.13 for ever-exposure and 1.32 for heavy-exposure, respectively [48]. However, a large international study of nearly 29,000 individuals from 34 countries (both high and low/middle income) found that working in settings with high exposure to dusts or fumes associated with increased respiratory symptoms, but did not correlate with decreases in postbronchodilator spirometry consistent with COPD [49].

Allergy and asthma

Airway responsiveness — Increased airway responsiveness to allergens or other external triggers is a risk factor for COPD, according to numerous observational studies [50-58]. As an example, one retrospective cohort study (n = 9651) found that the incidence of COPD over 11 years was higher among individuals with increased airway responsiveness, compared to those without increased airway responsiveness (OR 4.5, 95% CI 3.3-6.0) [55].

While it is certain that both airway responsiveness and smoking are independent risk factors for COPD, conflicting data make it unclear whether they interact. While several studies suggest that cigarette smoking increases the effect of airway responsiveness on the development of COPD [59], other studies do not [22,51-54].

Atopy — Atopy may increase an individual's risk for COPD, according to an observational study of 1025 older men (mean age 61 years) without asthma who underwent baseline skin prick and pulmonary function testing and were then followed for a median of three years [60]. Atopy was considered present when there was a mean wheal size ≥2 mm in response to four antigens: house dust, mixed grasses, mixed trees, and ragweed. Atopy predicted an excess annual rate of decline of the FEV1 (9.5 mL per year) and the FEV1/FVC (0.3 percent per year), compared with nonatopic patients. Animal studies provide further support that atopy and COPD are related [61].

Asthma — Mild to moderate persistent asthma is a risk factor for the development of COPD. In a study that utilized the Childhood Asthma Management Cohort (684 participants) to define clinical growth patterns in FEV1 over 18 years of follow-up, the FEV1 growth patterns of reduced growth, early decline, or both reduced growth and early decline were associated with the diagnosis of COPD based on spirometric criteria for airflow limitation (post-bronchodilator FEV1/FVC <0.70) [1,62]. Similarly, In a separate longitudinal population-based cohort with more than 45 years of follow-up, patients with asthma that remitted during childhood or adulthood had modest increases in risk for COPD based on post-bronchodilator airflow limitation in middle age (OR 2.0, 95% CI 1.1-3.6 and 3.6, 95% CI 1.3-10, respectively), whereas those with long-standing asthma had highly elevated risk of meeting these spirometric criteria for COPD (OR 8.7 and 6.7 for childhood-onset and adolescent/adult-onset persistent asthma, respectively) [63].

Abnormal lung development — Bronchopulmonary dysplasia, also known as neonatal chronic lung disease (CLD), is a consequence of preterm birth that is defined by dependence on supplemental oxygen for more than 28 days post-partum. Radiographic emphysema and evidence of airflow limitation on pulmonary function testing have been noted in young adult survivors of moderate and severe bronchopulmonary dysplasia [64,65]. (See "Complications and long-term pulmonary outcomes of bronchopulmonary dysplasia", section on 'Obstructive lung disease in adulthood'.)

Infections

Childhood pneumonias — Some patients without environmental exposures but with a history of prior infection demonstrate persistent airflow obstruction [4]. Infections in infancy, particularly respiratory syncytial virus, appear to demonstrate particularly increased risk [66,67]. The suspected mechanism is abnormal repair of injured lung leading to airway and parenchymal distortion in areas of prior infection.

Tuberculosis — Pulmonary tuberculosis may contribute to airflow obstruction via endobronchial infection and subsequent bronchostenosis or via lung parenchymal destruction with loss of airway tethering and is a risk factor for chronic airway obstruction in population studies [6,68]. In a study of 8784 Chinese subjects aged 50 or older, radiographic evidence of prior pulmonary tuberculosis was associated with an increased risk for airflow obstruction, independent of cigarette smoking, biomass fuel exposure, and prior diagnosis of asthma [69]. (See "Pulmonary tuberculosis: Clinical manifestations and complications", section on 'Endobronchial tuberculosis'.)

HIV — With an increase in worldwide use of highly effective antiretroviral therapy, there has been increasing recognition of longer-term complications of HIV infection. One meta-analysis of 11 prevalence studies found that patients living with HIV had an increased risk of developing COPD compared to non-HIV controls (OR 1.14, 95% CI 1.05-1.25) [70]. Prevalence in the population increased with the prevalence of detectable viral load, but not with CD4 count. However, even those who are virally suppressed are likely at higher risk for significant lung function decline over time than the general population, particularly in the setting of concomitant tobacco use [71].

Other risk factors

Sex — Women appear to be more susceptible to developing COPD and emphysema than men [72,73]. In a study that assessed the amount of emphysema by measuring lung attenuation on computed tomography, men and women had a similar amount of emphysema overall, but women had smoked a substantially lower number of pack years [73].

Antioxidant deficiency — There are limited data suggesting that a deficiency of antioxidant vitamins (eg, vitamins C and E) may be a risk factor for COPD [74-76]. In theory, a deficiency of antioxidant vitamins leaves the host unable to defend itself against the destructive effects of oxidative radicals, which derive from both exogenous sources (eg, cigarette smoke) and endogenous sources (eg, lung phagocytes).

As an example, in one prospective cohort study including smokers and former smokers, those in the highest quintile of antioxidant dietary intake (from flavonoids) had reduced risk of developing COPD compared with those in the lowest quintile, even after accounting for smoking exposure, physical activity, socioeconomic status, and other factors (hazard ratio 0.8) [77].

MOLECULAR RISK FACTORS — Observational studies indicate that molecular risk factors for chronic obstructive pulmonary disease (COPD) exist [22,78-81]. As an example, one observational study found that the risk of COPD was approximately three times higher among the first-degree relatives of patients who had severe premature COPD unrelated to alpha-1 antitrypsin deficiency [80]. The role of alpha-1 antitrypsin deficiency in COPD is discussed separately. (See "Clinical manifestations, diagnosis, and natural history of alpha-1 antitrypsin deficiency".)

Molecular risk factors for COPD have been assessed using several different methods, including studies of gene polymorphisms, antioxidant enzyme function, metalloproteinase dysregulation, and abnormalities that cause excess elastase.

Gene polymorphisms — Several gene polymorphisms (SNPs) have been identified that may increase the risk of COPD [82-95]. The functions of many of these genes are still unknown. Some examples are listed here:

Transforming growth factor beta 1 – Transforming growth factor beta is a member of a large superfamily of polypeptides involved in cellular growth, differentiation, and activation. SNPs of the gene encoding transforming growth factor beta 1 have been associated with development of COPD in smokers [85,88,89].

Serpine2 – Serpine2, also known as serpin peptidase inhibitor, was initially identified based on mouse and human fetal lung gene expression and then assessed in a case control study [96]. Serpine2 appears to be a COPD susceptibility gene that may be influenced by gene-by-smoking interaction.

Cystic fibrosis transmembrane conductance regulator (CFTR) – Loss of function of the CFTR gene causes cystic fibrosis (CF), but some combinations of deleterious variants can have incomplete penetrance or remain clinically silent. Assessment of coding variants for CFTR in cigarette smokers from two large cohort studies (COPDGene and ECLIPSE) found that the presence of a single CF-causing variant was associated with chronic bronchitis [97]. Additionally, the small number of subjects with two or more CFTR variants on separate alleles had significantly higher incidence of COPD than the cohort as a whole.

Other loci from genome-wide association studies – Genome-wide association studies (GWAS) have identified more than 80 separate loci associated with COPD or related disease. Notable loci include:

The earliest identified sites associated with COPD were 15q25 (CHRNA3/CHRN5/IREB2) [90], 4q31 (near HHIP) [91,92], and 4q22 (FAM13A) [93].

Notable additional loci associated with COPD were identified at 19q13 (near CYPA6, a locus also associated with cigarette smoking) [94], 5q32 (near the gene for 5-hydroxytryptamine receptor 4 [HTR4], a locus also associated with lung function in the general population) [86], as well as 6p21 (AGER) [98], and 14q32 (RIN3) [99].

Several more loci have been associated with chronic bronchitis (11p15.5 [EFCAB4A, CHID1, APA2A2]) [100] and airway hyper-responsiveness in COPD (9p21.2 near Lingo2, 3q13.1 near MyH15, and 5q33 near the sarcoglycan delta [SGCD] gene) [101]. While COPD lags behind asthma in terms of GWAS-identified loci are concerned, the gap is narrowing.

A separate GWAS was performed in a population based cohort (Multi-Ethnic Study of Atherosclerosis [MESA]-SNP Health Association Resource [SHARe]) in the United States and found two SNPs associated with the percentage of lung emphysema determined by computed tomography [102]. In addition, genes related to alpha-mannosidase appeared to be associated with the ratio of upper to lower lobe emphysema in some ethnic groups.

At least four of the loci associated with COPD overlap with idiopathic pulmonary fibrosis, with the two diseases having allelic effects in opposite directions, possibly indicating a molecular switch at that genetic locus [103].

A polygenic risk score developed using a lung function-based GWAS strongly associates with the likelihood of COPD in multiple cohorts of European ancestry [104], and higher polygenic risk scores may help identify patients at risk for early incident COPD [105].

Antioxidant related enzymes — Genetic variation in antioxidant enzyme function or regulation may affect risk for COPD [106]. In particular, the genes for glutathione S-transferases P1 and M1, glutamate cysteine ligase, and superoxide dismutase appear to be involved. Gene association studies are not available for some other antioxidant enzymes (eg, thioredoxin, gamma-glutamyl transferase) that may turn out to be important.

Glutathione S-transferases – Glutathione S-transferase P1 (GSTP1) aids in the detoxification of a number of substances that are found in cigarette smoke. Decreased glutathione S-transferase P1 activity due to genetic polymorphisms may increase the frequency of COPD [107-109]. Several case-control studies have identified a specific polymorphism in exon 5 (Ile105Val) that is more common among persons with COPD than controls [108,110]. Homozygous deletion of glutathione S-transferase M1 (GSTM1) has been associated with increased COPD risk in some, but not all studies [85,106,107,110].

Glutamate cysteine ligase – Glutamate cysteine ligase (GCL) is one of the three enzymes that relate to glutathione synthesis. Genetic variants in the promoter region and in the catalytic subunit that cause decreased glutathione levels have been associated with and increase risk of COPD [106,111].

Metalloproteinase dysregulation — Matrix metalloproteinases (MMPs) are a family of zinc-dependent enzymes that degrade extracellular matrix proteins. The activity of MMPs is regulated by tissue inhibitors of metalloproteinase (TIMPs). Numerous observational studies have demonstrated an association between COPD and abnormal activity of certain MMP or TIMP subtypes [112-116]:

A case-control study compared the bronchoalveolar lavage fluid of patients with emphysema to normal controls [113]. Patients with emphysema had increased MMP-1 (collagenase-1) expression and absent MMP-12 (macrophage elastase) activity [113].

Sputum from patients with asthma and COPD has increased MMP-2 (gelatinase A), MMP-9 (gelatinase B), MMP-8 (Collagenase 2), and TIMP-1 activity, compared to controls [114,116]. In addition, patients with stable COPD have an increased concentration of serum TIMP-1, compared to controls and patients with stable asthma [117].

MMP-12 has been identified as a gene associated with reduced lung function in asthma and early decline in lung function in COPD [118,119]. This gene has been associated with emphysema in mouse models of COPD [120].

The relationships among MMPs, TIMPs, and COPD are an area of ongoing research, with MMP inhibitors under investigation as therapeutic agents for COPD [112,121,122].

Excess elastase — The possibility that excess elastase contributes to COPD is suggested by two principal observations:

Premature emphysema is associated with deficiency of alpha-1 antitrypsin, an inhibitor of neutrophil elastase. (See "Clinical manifestations, diagnosis, and natural history of alpha-1 antitrypsin deficiency".)

Macrophage elastase-deficient mice do not develop emphysema despite intense, long-term exposure to cigarette smoke [120]. This is true even if excess inflammatory cells are attracted to the lung with exogenously administered monocyte chemoattractant protein-1.

RISK REDUCTION — Most of the clinical risk factors for chronic obstructive pulmonary disease (COPD) can be modified. However, it is difficult to directly measure the impact of risk factor modification on the incidence of COPD because of the extended duration between exposure to the risk factor and the onset of measurable airway obstruction. Studies need to be conducted over several decades for direct measurement.

As an alternative approach, most studies determine the rate of lung function decline and use it as an indirect measure of the risk of developing COPD. The goal of risk factor modification is to mitigate lung function decline, since patients with increased lung function decline are more likely to develop COPD. (See 'Lung function' above.)

Indirect evidence suggests that smoking cessation has the greatest impact on preventing COPD. Physical activity and avoidance of inhalational exposures may also reduce the incidence of COPD. Anti-inflammatory therapy and antioxidant therapy have been studied but appear to have minimal impact on the development of COPD.

Smoking cessation — Smoking cessation reduces the accelerated decline in lung function that is associated with smoking, which decreases the likelihood that COPD will develop [11,123,124].

This was illustrated by a retrospective cohort study (n = 8045) that found the incidence of COPD over 25 years was less among patients who had never smoked or quit smoking than among patients who continued to smoke [11]. Specifically, the incidence of COPD among never smokers, smokers who quit prior to the study, smokers who quit during the initial five years of the study, smokers who quit 5 to 15 years into the study, smokers who quit 15 to 25 years into the study, and those who continued to smoke was 4, 12, 5, 14, 24, and 41 percent, respectively among men. The incidence was 9, 11, 20, 25, 29, and 31 percent, respectively, among women.

Additional benefits of smoking cessation, as well as risks and approaches are discussed separately. (See "Benefits and consequences of smoking cessation" and "Overview of smoking cessation management in adults" and "Behavioral approaches to smoking cessation".)

Exposure avoidance — Reduction of environmental exposure to particulate matter, dusts, gases, vapors, fumes, or organic antigens is associated with slower lung function decline, but to a much smaller degree than smoking cessation.

This was demonstrated by a retrospective cohort study (n = 9651), which found that decreased particulate matter concentration was associated with a small reduction of the annual rate of decline of the forced expiratory volume in one second (FEV1) over 11 years [125]. Specifically, a 10 mcg per m3 annual decrease in the concentration of particulate matter was associated with a 3 mL reduction of the annual decrease of FEV1. This effect is small and of limited clinical relevance to individual patients, but may have public health relevance [126].

In a nine-year prospective cohort study, improved kitchen ventilation and/or use of biogas instead of biomass fuel were associated with a reduced decline in FEV1 [127]. When both interventions were utilized, the decline in FEV1 was decreased by 16 mL/year (95% CI 9-23 mL/year).

A variety of strategies are available to reduce the burden of inhaled particles, gases, vapors, and fumes [1,127]:

Implement, monitor, and enforce strict control of airborne exposure in the workplace

Initiate intensive and continuing education of workers, managers, clinicians, and legislators

Promote smoking cessation since smoking aggravates exposure to other particles and gases

Improve ventilation in areas where biomass fuels are used for cooking and promote use of clean fuels

Physical activity — Physical activity may mitigate lung function decline in active smokers. In a retrospective cohort study, 6790 volunteers were followed for a median duration of 11 years [128]. Active smokers with a moderate to high level of physical activity were less likely to develop COPD than active smokers with a low level of physical activity (OR 0.77, 95% CI 0.61-0.97). Additional studies are necessary before physical activity can be recommended as a means of decreasing the incidence of COPD.

Anti-inflammatory therapy — The observation that increased airway responsiveness is a risk factor for COPD led to the hypothesis that anti-inflammatory therapy may mitigate accelerated lung function decline.

Inhaled glucocorticoids – The use of inhaled glucocorticoids in young adults to prevent the onset of COPD has not been studied. However, the impact of inhaled glucocorticoids on lung function decline in patients with established COPD has been extensively studied and is discussed separately. (See "Role of inhaled glucocorticoid therapy in stable COPD".)

Statins – Statins (hydroxymethylglutaryl [HMG] CoA reductase inhibitors) are generally used for their lipid lowering characteristics, but also appear to have anti-inflammatory properties. In observational studies of COPD [129-131], statins have been associated with a lower rate of decline in pulmonary function, reduced rate and severity of exacerbations, rate of hospitalizations, and mortality. However, in a randomized trial that compared simvastatin with placebo in 885 patients with COPD, simvastatin did not attenuate lung function decline or reduce exacerbations. These studies are described in greater detail separately. (See "COPD exacerbations: Prognosis, discharge planning, and prevention", section on 'Ineffective interventions'.)

The effect of other anti-inflammatory therapies (eg, nonsteroidal anti-inflammatory drugs, systemic glucocorticoids) has not been studied in relevant patient populations.

(N) acetylcysteine — (N) acetylcysteine is a thiol derivative that has potential antioxidant and mucoactive effects. These effects have been explored in patients with COPD with conflicting results, and systematic reviews have found little or no benefit on the reduction in exacerbations or quality of life. (See "Role of mucoactive agents and secretion clearance techniques in COPD", section on 'Thiols and thiol derivatives'.)

The impact of antioxidant therapy (eg, (N) acetylcysteine) on lung function decline was studied in response to observations that antioxidant deficiency may be a risk factor for COPD. A trial that randomly assigned 50 patients with COPD to receive (N) acetylcysteine (600 mg/day) or placebo for three years found no between group difference in the annual rate of lung function decline [132]. Similarly, in a study that used a higher dose of (N) acetylcysteine (1200 mg/day) in 120 patients with COPD, no difference was found in the rate of decline in FEV1, although a slight reduction in the rate of exacerbations was noted in the (N) acetylcysteine group [133]. These studies are described in greater detail separately. (See "Role of mucoactive agents and secretion clearance techniques in COPD", section on 'Thiols and thiol derivatives'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Chronic obstructive pulmonary disease".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, “The Basics” and “Beyond the Basics.” The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on “patient info” and the keyword(s) of interest.)

Basics topics (see "Patient education: Chronic bronchitis (The Basics)")

Beyond the Basics topics (see "Patient education: Chronic obstructive pulmonary disease (COPD) (Beyond the Basics)" and "Patient education: Chronic obstructive pulmonary disease (COPD) treatments (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

COPD results from complex interactions between clinical and molecular (ie, genetic) risk factors. (See 'Introduction' above.)

Definite or possible risk factors for COPD include inhalational exposure (eg, smoking), increased airway responsiveness, atopy, and antioxidant deficiency. (See 'Clinical risk factors' above.)

Molecular risk factors for COPD include a variety of gene polymorphisms, antioxidant related enzyme dysfunction, metalloproteinase dysregulation, and abnormalities that cause excess elastase. (See 'Molecular risk factors' above.)

Some of the clinical risk factors for COPD can be modified, thereby reducing the rate of lung function decline. (See 'Risk reduction' above.)

Smoking cessation is the most important way to reduce the rate of lung function decline and the risk of developing COPD among those who smoke. (See 'Smoking cessation' above.)

Whenever possible, environmental exposure to particulate matter, dusts, gases, vapors, and fumes should be avoided or reduced. (See 'Exposure avoidance' above.)

Physical activity may mitigate lung function decline, although further studies are needed. (See 'Physical activity' above.)

  1. Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global Strategy for the Diagnosis, Management and Prevention of Chronic Obstructive Pulmonary Disease: 2024 Report. www.goldcopd.org www.goldcopd.org (Accessed on November 16, 2023).
  2. Postma DS, Bush A, van den Berge M. Risk factors and early origins of chronic obstructive pulmonary disease. Lancet 2015; 385:899.
  3. Eisner MD, Anthonisen N, Coultas D, et al. An official American Thoracic Society public policy statement: Novel risk factors and the global burden of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2010; 182:693.
  4. de Marco R, Accordini S, Marcon A, et al. Risk factors for chronic obstructive pulmonary disease in a European cohort of young adults. Am J Respir Crit Care Med 2011; 183:891.
  5. Perret JL, Dharmage SC, Matheson MC, et al. The interplay between the effects of lifetime asthma, smoking, and atopy on fixed airflow obstruction in middle age. Am J Respir Crit Care Med 2013; 187:42.
  6. Burney P, Patel J, Minelli C, et al. Prevalence and Population-Attributable Risk for Chronic Airflow Obstruction in a Large Multinational Study. Am J Respir Crit Care Med 2021; 203:1353.
  7. Celli B, Fabbri L, Criner G, et al. Definition and Nomenclature of Chronic Obstructive Pulmonary Disease: Time for Its Revision. Am J Respir Crit Care Med 2022; 206:1317.
  8. Tager IB, Speizer FE. Risk estimates for chronic bronchitis in smokers: a study of male-female differences. Am Rev Respir Dis 1976; 113:619.
  9. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking, changes in smoking habits, and rate of decline in FEV1: new insight into gender differences. Eur Respir J 1994; 7:1056.
  10. Doll R, Peto R. Mortality in relation to smoking: 20 years' observations on male British doctors. Br Med J 1976; 2:1525.
  11. Løkke A, Lange P, Scharling H, et al. Developing COPD: a 25 year follow up study of the general population. Thorax 2006; 61:935.
  12. van Durme YMTA, Verhamme KMC, Stijnen T, et al. Prevalence, incidence, and lifetime risk for the development of COPD in the elderly: the Rotterdam study. Chest 2009; 135:368.
  13. Wang C, Xu J, Yang L, et al. Prevalence and risk factors of chronic obstructive pulmonary disease in China (the China Pulmonary Health [CPH] study): a national cross-sectional study. Lancet 2018; 391:1706.
  14. US Burden of Disease Collaborators, Mokdad AH, Ballestros K, et al. The State of US Health, 1990-2016: Burden of Diseases, Injuries, and Risk Factors Among US States. JAMA 2018; 319:1444.
  15. 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; 155:179.
  16. Simel D, Rennie D. The rational clinical examination: Evidence-based clinical diagnosis, McGraw Hill (Ed), New York 2008.
  17. Bhatt SP, Kim YI, Harrington KF, et al. Smoking duration alone provides stronger risk estimates of chronic obstructive pulmonary disease than pack-years. Thorax 2018; 73:414.
  18. Sargent JD, Halenar M, Steinberg AW, et al. Childhood Cigarette Smoking and Risk of Chronic Obstructive Pulmonary Disease in Older U.S. Adults. Am J Respir Crit Care Med 2023; 208:428.
  19. McKleroy W, Shing T, Anderson WH, et al. Longitudinal Follow-Up of Participants With Tobacco Exposure and Preserved Spirometry. JAMA 2023; 330:442.
  20. She J, Yang P, Wang Y, et al. Chinese water-pipe smoking and the risk of COPD. Chest 2014; 146:924.
  21. Tan WC, Lo C, Jong A, et al. Marijuana and chronic obstructive lung disease: a population-based study. CMAJ 2009; 180:814.
  22. Celedon JC, Speizer FE, Drazen JM, et al. Bronchodilator responsiveness and serum total IgE levels in families of probands with severe early-onset COPD. Eur Respir J 1999; 14:1009.
  23. Korn RJ, Dockery DW, Speizer FE, et al. Occupational exposures and chronic respiratory symptoms. A population-based study. Am Rev Respir Dis 1987; 136:298.
  24. Oxman AD, Muir DC, Shannon HS, et al. Occupational dust exposure and chronic obstructive pulmonary disease. A systematic overview of the evidence. Am Rev Respir Dis 1993; 148:38.
  25. Sunyer J, Zock JP, Kromhout H, et al. Lung function decline, chronic bronchitis, and occupational exposures in young adults. Am J Respir Crit Care Med 2005; 172:1139.
  26. Terho EO. Work-related respiratory disorders among Finnish farmers. Am J Ind Med 1990; 18:269.
  27. Dalphin JC, Debieuvre D, Pernet D, et al. Prevalence and risk factors for chronic bronchitis and farmer's lung in French dairy farmers. Br J Ind Med 1993; 50:941.
  28. Terho EO, Husman K, Vohlonen I. Prevalence and incidence of chronic bronchitis and farmer's lung with respect to age, sex, atopy, and smoking. Eur J Respir Dis Suppl 1987; 152:19.
  29. Torres-Duque C, Maldonado D, Pérez-Padilla R, et al. Biomass fuels and respiratory diseases: a review of the evidence. Proc Am Thorac Soc 2008; 5:577.
  30. Pope CA 3rd, Ezzati M, Dockery DW. Fine-particulate air pollution and life expectancy in the United States. N Engl J Med 2009; 360:376.
  31. Zhou Y, Wang C, Yao W, et al. COPD in Chinese nonsmokers. Eur Respir J 2009; 33:509.
  32. Hu G, Zhou Y, Tian J, et al. Risk of COPD from exposure to biomass smoke: a metaanalysis. Chest 2010; 138:20.
  33. Andersen ZJ, Hvidberg M, Jensen SS, 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; 183:455.
  34. Mehta AJ, Miedinger D, Keidel D, et al. Occupational exposure to dusts, gases, and fumes and incidence of chronic obstructive pulmonary disease in the Swiss Cohort Study on Air Pollution and Lung and Heart Diseases in Adults. Am J Respir Crit Care Med 2012; 185:1292.
  35. Guillien A, Puyraveau M, Soumagne T, et al. Prevalence and risk factors for COPD in farmers: a cross-sectional controlled study. Eur Respir J 2016; 47:95.
  36. Borlée F, Yzermans CJ, Aalders B, et al. Air Pollution from Livestock Farms Is Associated with Airway Obstruction in Neighboring Residents. Am J Respir Crit Care Med 2017; 196:1152.
  37. Syamlal G, Kurth LM, Dodd KE, et al. Chronic Obstructive Pulmonary Disease Mortality by Industry and Occupation - United States, 2020. MMWR Morb Mortal Wkly Rep 2022; 71:1550.
  38. Xing Z, Yang T, Shi S, et al. Combined effect of ozone and household air pollution on COPD in people aged less than 50 years old. Thorax 2023; 79:35.
  39. Hansel NN, Woo H, Koehler K, et al. Indoor Pollution and Lung Function Decline in Current and Former Smokers: SPIROMICS AIR. Am J Respir Crit Care Med 2023; 208:1042.
  40. Kurmi OP, Semple S, Simkhada P, et al. COPD and chronic bronchitis risk of indoor air pollution from solid fuel: a systematic review and meta-analysis. Thorax 2010; 65:221.
  41. Po JY, FitzGerald JM, Carlsten C. Respiratory disease associated with solid biomass fuel exposure in rural women and children: systematic review and meta-analysis. Thorax 2011; 66:232.
  42. Sin DD, Doiron D, Agusti A, et al. Air pollution and COPD: GOLD 2023 committee report. Eur Respir J 2023; 61.
  43. Park J, Kim HJ, Lee CH, et al. Impact of long-term exposure to ambient air pollution on the incidence of chronic obstructive pulmonary disease: A systematic review and meta-analysis. Environ Res 2021; 194:110703.
  44. Doiron D, de Hoogh K, Probst-Hensch N, et al. Air pollution, lung function and COPD: results from the population-based UK Biobank study. Eur Respir J 2019; 54.
  45. Shin S, Bai L, Burnett RT, et al. Air Pollution as a Risk Factor for Incident Chronic Obstructive Pulmonary Disease and Asthma. A 15-Year Population-based Cohort Study. Am J Respir Crit Care Med 2021; 203:1138.
  46. Schikowski T, Adam M, Marcon A, et al. Association of ambient air pollution with the prevalence and incidence of COPD. Eur Respir J 2014; 44:614.
  47. Dumas O, Varraso R, Boggs KM, et al. Association of Occupational Exposure to Disinfectants With Incidence of Chronic Obstructive Pulmonary Disease Among US Female Nurses. JAMA Netw Open 2019; 2:e1913563.
  48. De Matteis S, Jarvis D, Darnton L, et al. Lifetime occupational exposures and chronic obstructive pulmonary disease risk in the UK Biobank cohort. Thorax 2022; 77:997.
  49. Ratanachina J, Amaral AFS, De Matteis S, et al. Association of respiratory symptoms and lung function with occupation in the multinational Burden of Obstructive Lung Disease (BOLD) study. Eur Respir J 2023; 61.
  50. Silva GE, Sherrill DL, Guerra S, Barbee RA. Asthma as a risk factor for COPD in a longitudinal study. Chest 2004; 126:59.
  51. Frew AJ, Kennedy SM, Chan-Yeung M. Methacholine responsiveness, smoking, and atopy as risk factors for accelerated FEV1 decline in male working populations. Am Rev Respir Dis 1992; 146:878.
  52. O'Connor GT, Sparrow D, Weiss ST. A prospective longitudinal study of methacholine airway responsiveness as a predictor of pulmonary-function decline: the Normative Aging Study. Am J Respir Crit Care Med 1995; 152:87.
  53. Rijcken B, Schouten JP, Xu X, et al. Airway hyperresponsiveness to histamine associated with accelerated decline in FEV1. Am J Respir Crit Care Med 1995; 151:1377.
  54. Tracey M, Villar A, Dow L, et al. The influence of increased bronchial responsiveness, atopy, and serum IgE on decline in FEV1. A longitudinal study in the elderly. Am J Respir Crit Care Med 1995; 151:656.
  55. Brutsche MH, Downs SH, Schindler C, et al. Bronchial hyperresponsiveness and the development of asthma and COPD in asymptomatic individuals: SAPALDIA cohort study. Thorax 2006; 61:671.
  56. O'Connor GT, Sparrow D, Weiss ST. The role of allergy and nonspecific airway hyperresponsiveness in the pathogenesis of chronic obstructive pulmonary disease. Am Rev Respir Dis 1989; 140:225.
  57. Agnati LF, Zoli M, Biagini G, Fuxe K. Neuronal plasticity and ageing processes in the frame of the 'Red Queen Theory'. Acta Physiol Scand 1992; 145:301.
  58. Marcon A, Locatelli F, Keidel D, et al. Airway responsiveness to methacholine and incidence of COPD: an international prospective cohort study. Thorax 2018; 73:825.
  59. Aanerud M, Carsin AE, Sunyer J, et al. Interaction between asthma and smoking increases the risk of adult airway obstruction. Eur Respir J 2015; 45:635.
  60. Gottlieb DJ, Sparrow D, O'Connor GT, Weiss ST. Skin test reactivity to common aeroallergens and decline of lung function. The Normative Aging Study. Am J Respir Crit Care Med 1996; 153:561.
  61. Zheng T, Zhu Z, Wang Z, et al. Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema. J Clin Invest 2000; 106:1081.
  62. McGeachie MJ, Yates KP, Zhou X, et al. Patterns of Growth and Decline in Lung Function in Persistent Childhood Asthma. N Engl J Med 2016; 374:1842.
  63. Tan DJ, Lodge CJ, Walters EH, et al. Longitudinal Asthma Phenotypes from Childhood to Middle-Age: A Population-based Cohort Study. Am J Respir Crit Care Med 2023; 208:132.
  64. Wong PM, Lees AN, Louw J, et al. Emphysema in young adult survivors of moderate-to-severe bronchopulmonary dysplasia. Eur Respir J 2008; 32:321.
  65. Bårdsen T, Røksund OD, Eagan TML, et al. Impaired Lung Function in Extremely Preterm-Born Adults in Their Fourth Decade of Life. Am J Respir Crit Care Med 2023; 208:493.
  66. Baraldi E, Bonadies L, Manzoni P. Evidence on the Link between Respiratory Syncytial Virus Infection in Early Life and Chronic Obstructive Lung Diseases. Am J Perinatol 2020; 37:S26.
  67. Allinson JP, Chaturvedi N, Wong A, et al. Early childhood lower respiratory tract infection and premature adult death from respiratory disease in Great Britain: a national birth cohort study. Lancet 2023; 401:1183.
  68. Kamenar K, Hossen S, Gupte AN, et al. Previous tuberculosis disease as a risk factor for chronic obstructive pulmonary disease: a cross-sectional analysis of multicountry, population-based studies. Thorax 2022; 77:1088.
  69. Lam KB, Jiang CQ, Jordan RE, et al. Prior TB, smoking, and airflow obstruction: a cross-sectional analysis of the Guangzhou Biobank Cohort Study. Chest 2010; 137:593.
  70. Bigna JJ, Kenne AM, Asangbeh SL, Sibetcheu AT. Prevalence of chronic obstructive pulmonary disease in the global population with HIV: a systematic review and meta-analysis. Lancet Glob Health 2018; 6:e193.
  71. Thudium RF, Ronit A, Afzal S, et al. Faster lung function decline in people living with HIV despite adequate treatment: a longitudinal matched cohort study. Thorax 2023; 78:535.
  72. Kamil F, Pinzon I, Foreman MG. Sex and race factors in early-onset COPD. Curr Opin Pulm Med 2013; 19:140.
  73. Hardin M, Foreman M, Dransfield MT, et al. Sex-specific features of emphysema among current and former smokers with COPD. Eur Respir J 2016; 47:104.
  74. Cantin A, Crystal RG. Oxidants, antioxidants and the pathogenesis of emphysema. Eur J Respir Dis Suppl 1985; 139:7.
  75. Sanguinetti CM. Oxidant/antioxidant imbalance: role in the pathogenesis of COPD. Respiration 1992; 59 Suppl 1:20.
  76. Nowak D, Piasecka G, Antczak A, Pietras T. Effect of ascorbic acid on hydroxyl radical generation by chemical, enzymatic and cellular systems. Importance for antioxidant prevention of pulmonary emphysema. Biomed Biochim Acta 1991; 50:265.
  77. Bondonno NP, Parmenter BH, Dalgaard F, et al. Flavonoid intakes inversely associate with COPD in smokers. Eur Respir J 2022; 60.
  78. Barnes PJ. Genetics and pulmonary medicine. 9. Molecular genetics of chronic obstructive pulmonary disease. Thorax 1999; 54:245.
  79. Silverman EK. Genetics of chronic obstructive pulmonary disease. Novartis Found Symp 2001; 234:45.
  80. Silverman EK, Chapman HA, Drazen JM, et al. Genetic epidemiology of severe, early-onset chronic obstructive pulmonary disease. Risk to relatives for airflow obstruction and chronic bronchitis. Am J Respir Crit Care Med 1998; 157:1770.
  81. Marciniak SJ, Lomas DA. What can naturally occurring mutations tell us about the pathogenesis of COPD? Thorax 2009; 64:359.
  82. Silverman EK, Mosley JD, Palmer LJ, et al. Genome-wide linkage analysis of severe, early-onset chronic obstructive pulmonary disease: airflow obstruction and chronic bronchitis phenotypes. Hum Mol Genet 2002; 11:623.
  83. Silverman EK, Palmer LJ, Mosley JD, et al. Genomewide linkage analysis of quantitative spirometric phenotypes in severe early-onset chronic obstructive pulmonary disease. Am J Hum Genet 2002; 70:1229.
  84. Palmer LJ, Celedón JC, Chapman HA, et al. Genome-wide linkage analysis of bronchodilator responsiveness and post-bronchodilator spirometric phenotypes in chronic obstructive pulmonary disease. Hum Mol Genet 2003; 12:1199.
  85. Smolonska J, Wijmenga C, Postma DS, Boezen HM. Meta-analyses on suspected chronic obstructive pulmonary disease genes: a summary of 20 years' research. Am J Respir Crit Care Med 2009; 180:618.
  86. Wilk JB, Shrine NR, Loehr LR, et al. Genome-wide association studies identify CHRNA5/3 and HTR4 in the development of airflow obstruction. Am J Respir Crit Care Med 2012; 186:622.
  87. Soler Artigas M, Loth DW, Wain LV, et al. Genome-wide association and large-scale follow up identifies 16 new loci influencing lung function. Nat Genet 2011; 43:1082.
  88. Celedón JC, Lange C, Raby BA, et al. The transforming growth factor-beta1 (TGFB1) gene is associated with chronic obstructive pulmonary disease (COPD). Hum Mol Genet 2004; 13:1649.
  89. Wu L, Chau J, Young RP, et al. Transforming growth factor-beta1 genotype and susceptibility to chronic obstructive pulmonary disease. Thorax 2004; 59:126.
  90. DeMeo DL, Mariani T, Bhattacharya S, et al. Integration of genomic and genetic approaches implicates IREB2 as a COPD susceptibility gene. Am J Hum Genet 2009; 85:493.
  91. Pillai SG, Kong X, Edwards LD, et al. Loci identified by genome-wide association studies influence different disease-related phenotypes in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2010; 182:1498.
  92. Zhou X, Baron RM, Hardin M, et al. Identification of a chronic obstructive pulmonary disease genetic determinant that regulates HHIP. Hum Mol Genet 2012; 21:1325.
  93. Cho MH, Boutaoui N, Klanderman BJ, et al. Variants in FAM13A are associated with chronic obstructive pulmonary disease. Nat Genet 2010; 42:200.
  94. Cho MH, Castaldi PJ, Wan ES, et al. A genome-wide association study of COPD identifies a susceptibility locus on chromosome 19q13. Hum Mol Genet 2012; 21:947.
  95. Kim W, Prokopenko D, Sakornsakolpat P, et al. Genome-Wide Gene-by-Smoking Interaction Study of Chronic Obstructive Pulmonary Disease. Am J Epidemiol 2021; 190:875.
  96. Demeo DL, Mariani TJ, Lange C, et al. The SERPINE2 gene is associated with chronic obstructive pulmonary disease. Am J Hum Genet 2006; 78:253.
  97. Saferali A, Qiao D, Kim W, et al. CFTR variants are associated with chronic bronchitis in smokers. Eur Respir J 2022; 60.
  98. Cho MH, Castaldi PJ, Hersh CP, et al. A Genome-Wide Association Study of Emphysema and Airway Quantitative Imaging Phenotypes. Am J Respir Crit Care Med 2015; 192:559.
  99. Lutz SM, Cho MH, Young K, et al. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet 2015; 16:138.
  100. Lee JH, Cho MH, Hersh CP, et al. Genetic susceptibility for chronic bronchitis in chronic obstructive pulmonary disease. Respir Res 2014; 15:113.
  101. Hansel NN, Paré PD, Rafaels N, et al. Genome-Wide Association Study Identification of Novel Loci Associated with Airway Responsiveness in Chronic Obstructive Pulmonary Disease. Am J Respir Cell Mol Biol 2015; 53:226.
  102. Manichaikul A, Hoffman EA, Smolonska J, et al. Genome-wide study of percent emphysema on computed tomography in the general population. The Multi-Ethnic Study of Atherosclerosis Lung/SNP Health Association Resource Study. Am J Respir Crit Care Med 2014; 189:408.
  103. Sakornsakolpat P, Prokopenko D, Lamontagne M, et al. Genetic landscape of chronic obstructive pulmonary disease identifies heterogeneous cell-type and phenotype associations. Nat Genet 2019; 51:494.
  104. Moll M, Sakornsakolpat P, Shrine N, et al. Chronic obstructive pulmonary disease and related phenotypes: polygenic risk scores in population-based and case-control cohorts. Lancet Respir Med 2020; 8:696.
  105. Zhang J, Xu H, Qiao D, et al. A polygenic risk score and age of diagnosis of COPD. Eur Respir J 2022; 60.
  106. Bentley AR, Emrani P, Cassano PA. Genetic variation and gene expression in antioxidant related enzymes and risk of COPD: a systematic review. Thorax 2008; 63:956.
  107. Cheng SL, Yu CJ, Chen CJ, Yang PC. Genetic polymorphism of epoxide hydrolase and glutathione S-transferase in COPD. Eur Respir J 2004; 23:818.
  108. Ishii T, Matsuse T, Teramoto S, et al. Glutathione S-transferase P1 (GSTP1) polymorphism in patients with chronic obstructive pulmonary disease. Thorax 1999; 54:693.
  109. Yim JJ, Yoo CG, Lee CT, et al. Lack of association between glutathione S-transferase P1 polymorphism and COPD in Koreans. Lung 2002; 180:119.
  110. He JQ, Connett JE, Anthonisen NR, et al. Glutathione S-transferase variants and their interaction with smoking on lung function. Am J Respir Crit Care Med 2004; 170:388.
  111. Liu S, Li B, Zhou Y, et al. Genetic analysis of CC16, OGG1 and GCLC polymorphisms and susceptibility to COPD. Respirology 2007; 12:29.
  112. Churg A, Wright JL. Proteases and emphysema. Curr Opin Pulm Med 2005; 11:153.
  113. Imai K, Dalal SS, Chen ES, et al. Human collagenase (matrix metalloproteinase-1) expression in the lungs of patients with emphysema. Am J Respir Crit Care Med 2001; 163:786.
  114. Cataldo D, Munaut C, Noël A, et al. MMP-2- and MMP-9-linked gelatinolytic activity in the sputum from patients with asthma and chronic obstructive pulmonary disease. Int Arch Allergy Immunol 2000; 123:259.
  115. Leco KJ, Waterhouse P, Sanchez OH, et al. Spontaneous air space enlargement in the lungs of mice lacking tissue inhibitor of metalloproteinases-3 (TIMP-3). J Clin Invest 2001; 108:817.
  116. Vernooy JH, Lindeman JH, Jacobs JA, et al. Increased activity of matrix metalloproteinase-8 and matrix metalloproteinase-9 in induced sputum from patients with COPD. Chest 2004; 126:1802.
  117. Higashimoto Y, Yamagata Y, Iwata T, et al. Increased serum concentrations of tissue inhibitor of metalloproteinase-1 in COPD patients. Eur Respir J 2005; 25:885.
  118. Hunninghake GM, Cho MH, Tesfaigzi Y, et al. MMP12, lung function, and COPD in high-risk populations. N Engl J Med 2009; 361:2599.
  119. Ngo D, Pratte KA, Flexeder C, et al. Systemic Markers of Lung Function and Forced Expiratory Volume in 1 Second Decline across Diverse Cohorts. Ann Am Thorac Soc 2023; 20:1124.
  120. Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science 1997; 277:2002.
  121. Mao JT, Tashkin DP, Belloni PN, et al. All-trans retinoic acid modulates the balance of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in patients with emphysema. Chest 2003; 124:1724.
  122. Cawston T, Carrere S, Catterall J, et al. Matrix metalloproteinases and TIMPs: properties and implications for the treatment of chronic obstructive pulmonary disease. Novartis Found Symp 2001; 234:205.
  123. Postma DS, Burema J, Gimeno F, et al. Prognosis in severe chronic obstructive pulmonary disease. Am Rev Respir Dis 1979; 119:357.
  124. Anthonisen NR, Connett JE, Kiley JP, et al. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA 1994; 272:1497.
  125. Downs SH, Schindler C, Liu LJ, et al. Reduced exposure to PM10 and attenuated age-related decline in lung function. N Engl J Med 2007; 357:2338.
  126. Lippmann M. Health effects of airborne particulate matter. N Engl J Med 2007; 357:2395.
  127. Zhou Y, Zou Y, Li X, et al. Lung Function and Incidence of Chronic Obstructive Pulmonary Disease after Improved Cooking Fuels and Kitchen Ventilation: A 9-Year Prospective Cohort Study. PLoS Med 2014; 11.
  128. Garcia-Aymerich J, Lange P, Benet M, et al. Regular physical activity modifies smoking-related lung function decline and reduces risk of chronic obstructive pulmonary disease: a population-based cohort study. Am J Respir Crit Care Med 2007; 175:458.
  129. Keddissi JI, Younis WG, Chbeir EA, et al. The use of statins and lung function in current and former smokers. Chest 2007; 132:1764.
  130. Janda S, Park K, FitzGerald JM, et al. Statins in COPD: a systematic review. Chest 2009; 136:734.
  131. Wang MT, Lo YW, Tsai CL, et al. Statin use and risk of COPD exacerbation requiring hospitalization. Am J Med 2013; 126:598.
  132. Decramer M, Rutten-van Mölken M, Dekhuijzen PN, et al. Effects of N-acetylcysteine on outcomes in chronic obstructive pulmonary disease (Bronchitis Randomized on NAC Cost-Utility Study, BRONCUS): a randomised placebo-controlled trial. Lancet 2005; 365:1552.
  133. Tse HN, Raiteri L, Wong KY, et al. High-dose N-acetylcysteine in stable COPD: the 1-year, double-blind, randomized, placebo-controlled HIACE study. Chest 2013; 144:106.
Topic 1443 Version 58.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟