Jump to content

Chronic obstructive pulmonary disease

This is a good article. Click here for more information.
From Wikipedia, the free encyclopedia
(Redirected fromCOPD)

Chronic obstructive pulmonary disease
Other namesChronic obstructive lung disease (COLD), chronic obstructive airway disease (COAD)
Section of a lung showingcentrilobular emphysema,with enlarged airspaces in the centre of alobuleusually caused by smoking and a major feature of COPD
SpecialtyPulmonology
SymptomsShortness of breath,chroniccough[1]
ComplicationsAnxiety,depression,pulmonary heart disease,pneumothorax[2][1]
Usual onsetOver 35 years old[1]
DurationLong term[1]
CausesTobacco smoking,air pollution,genetics[3]
Diagnostic methodSpirometry[4]
Differential diagnosisAsthma,congestive heart failure,bronchiectasis,tuberculosis,obliterative bronchiolitis,diffuse panbronchiolitis[5]
PreventionStopping smoking,improving indoor and outdoorair quality,tobacco controlmeasures[3][6]
TreatmentPulmonary rehabilitation,long-term oxygen therapy,lung volume reduction,[6]
MedicationInhaledbronchodilatorsandsteroids[6]
Frequency174.5 million (2015)[7]
Deaths3.2 million (2019)[3]

Chronic obstructive pulmonary disease(COPD) is a type of progressivelung diseasecharacterized by long-term respiratory symptoms and airflow limitation.[8]GOLD2024 defined COPD as a heterogeneous lung condition characterized by chronic respiratory symptoms (dyspneaor shortness of breath, cough, sputum production and/or exacerbations) due to abnormalities of the airways (bronchitis,bronchiolitis) and/oralveoli(emphysema) that cause persistent, often progressive, airflow obstruction.[9]

The main symptoms of COPD include shortness of breath and acough,which may or may not producemucus.[4]COPDprogressively worsens,witheveryday activitiessuch as walking or dressing becoming difficult.[3]While COPD is incurable, it is preventable and treatable. The two most common types of COPD areemphysemaandchronic bronchitisand have been the two classic COPDphenotypes.However, this basic dogma has been challenged as varying degrees of co-existing emphysema, chronic bronchitis, and potentially significant vascular diseases have all been acknowledged in those with COPD, giving rise to the classification of other phenotypes or subtypes.[10]

Emphysema is defined as enlarged airspaces (alveoli) whosewallshave broken down resulting in permanent damage to thelung tissue.Chronic bronchitis is defined as a productive cough that is present for at least three months each year for two years. Both of these conditions can exist without airflow limitation when they are not classed as COPD. Emphysema is just one of the structural abnormalities that can limit airflow and can exist without airflow limitation in a significant number of people.[11][12]Chronic bronchitis does not always result in airflow limitation. However, in young adults with chronic bronchitis who smoke, the risk of developing COPD is high.[13]Many definitions of COPD in the past included emphysema and chronic bronchitis, but these have never been included inGOLD reportdefinitions.[8]Emphysema and chronic bronchitis remain the predominant phenotypes of COPD but there is often overlap between them and a number of other phenotypes have also been described.[10][14]COPD andasthmamay coexist and converge in some individuals.[15]COPD is associated with low-grade systemic inflammation.[16]

The most common cause of COPD istobacco smoking.[17]Otherrisk factorsincludeindoorandoutdoor air pollutionincludingdust,exposure to occupational irritants such as dust fromgrains,cadmium dust or fumes,andgenetics,such asAlpha -1 antitrypsin deficiency.[13][18]Indeveloping countries,common sources ofhousehold air pollutionare the use of coal andbiomasssuch as wood anddry dungas fuel forcooking and heating.[19][13]The diagnosis is based on poor airflow as measured byspirometry.[4]

Most cases of COPD can be prevented by reducing exposure to risk factors such as smoking and indoor and outdoor pollutants.[20]While treatment can slow worsening, there is no conclusive evidence that any medications can change the long-term decline in lung function.[6]COPD treatments includesmoking cessation,vaccinations,pulmonary rehabilitation,inhaledbronchodilatorsandcorticosteroids.[6]Some people may benefit from long-termoxygen therapy,lung volume reductionandlung transplantation.[21]In those who have periods ofacute worsening,increased use of medications,antibiotics,corticosteroids and hospitalization may be needed.[22]

As of 2015, COPD affected about 174.5 million people (2.4% of the global population).[7]It typically occurs in males and females over the age of 35–40.[1][3]In 2019 it caused 3.2 million deaths, 80% occurring in lower and middle income countries,[3]up from 2.4 million deaths in 1990.[23][24]In 2021, it was the fourth biggest cause of death, responsible for approximately 5% of total deaths.[25]The number of deaths is projected to increase further because of continued exposure to risk factors and an aging population.[8]In the United States in 2010 the economic cost was put at US$32.1 billion and projected to rise to US$49 billion in 2020.[26]In the United Kingdom this cost is estimated at £3.8 billion annually.[27]

Signs and symptoms

[edit]
Signs and symptomsof stages of COPD

Shortness of breath

[edit]

Acardinal symptomof COPD is the chronic and progressiveshortness of breathwhich is most characteristic of the condition. Shortness of breath (breathlessness) is often the most distressing symptom responsible for the associated anxiety and level of disability experienced.[4]Symptoms ofwheezingand chest tightness associated with breathlessness can be variable over the course of a day or between days and are not always present. Chest tightness often follows exertion.[4]Many people with more advanced COPDbreathe through pursed lips,which can improve shortness of breath.[28]Shortness of breath is often responsible for reduced physical activity and low levels of physical activity are associated with worse outcomes.[29][30]In severe and very severe cases there may beconstant tiredness,weight loss, muscle loss andanorexia.People with COPD often have increased breathlessness and frequentcoldsbefore seeking treatment.[4]

Cough

[edit]

The most often first symptom of COPD is a chronic cough, which may or may not be productive ofmucusasphlegm.Phlegm coughed up assputumcan be intermittent and may be swallowed or spat out depending on social or cultural factors and is therefore not always easy to evaluate. However, an accompanying productive cough is only seen in up to 30% of cases. Sometimes limited airflow may develop in the absence of a cough.[4]Symptoms are usually worse in the morning.[31]

A chronicproductive coughis the result ofmucus hypersecretionand when it persists for more than three months each year for at least two years, it is defined aschronic bronchitis.[13]Chronic bronchitis can occur before the restricted airflow diagnostic of COPD.[8]Some people with COPD attribute the symptoms to the consequences of smoking. In severe COPD, vigorous coughing may lead torib fracturesor toa brief loss of consciousness.[4]

Exacerbations

[edit]

Anacute exacerbationis a sudden worsening ofsigns and symptomsthat lasts for several days. The key symptom is increased breathlessness, other more pronounced symptoms are of excessive mucus, increased cough and wheeze. A commonly found sign isair trappinggiving a difficulty in completeexhalation.[32] The usual cause of an exacerbation is aviral infection,most often thecommon cold.[13]The common cold is usually associated with the winter months but can occur at any time.[33]Otherrespiratory infectionsmay be bacterial or in combination sometimes secondary to a viral infection.[34]The most common bacterial infection is caused byHaemophilus influenzae.[35]Other risks include exposure to tobacco smoke (active andpassive) and environmentalpollutants– both indoor and outdoor.[36]During theCOVID-19 pandemic,hospital admissions for COPD exacerbations sharply decreased which may be attributable to reduction of emissions and cleaner air.[37]There has also been a marked decrease in the number of cold and flu infections during this time.[38]

Smoke from wildfiresis proving an increasing risk in many parts of the world and government agencies have published protective advice on their websites. In the US theEPAadvises that the use ofdust masksdo not give protection from the fine particles inwildfiresand instead advise the use of well-fittingparticulate masks.[39]This same advice is offered in Canada and Australia to the effects of their forest fires.[40][41]

The number of exacerbations is not seen to relate to any stage of the disease; those with two or more a year are classed asfrequent exacerbatorsand these lead to a worsening in the disease progression.[32]Frailtyin ageing increases exacerbations and hospitalization.[42]

Acute exacerbations in COPD are often unexplained and thought to have many causes other than infections. A study has emphasized the possibility of apulmonary embolismas sometimes being responsible in these cases. Signs can includepleuriticchest painandheart failurewithout signs of infection. Such emboli could respond toanticoagulants.[43]

Other conditions

[edit]

COPD often occurs along with a number of other conditions (comorbidities) due in part to shared risk factors. Common comorbidities includecardiovascular disease,skeletal muscledysfunction,metabolic syndrome,osteoporosis,depression,anxiety,asthmaandlung cancer.[44]Alpha-1 antitrypsin deficiency(A1AD) is an important risk factor for COPD.[45]It is advised that everybody with COPD be screened for A1AD.[46]Metabolic syndromehas been seen to affect up to fifty percent of those with COPD and significantly affects the outcomes.[47]When comorbid with COPD there is more systemic inflammation.[47]It is not known if it co-exists with COPD or develops as a consequence of the pathology. Metabolic syndrome on its own has a high rate of morbidity and mortality and this rate is amplified when comorbid with COPD.Tuberculosisis a risk factor for the development of COPD, and is also a potential comorbidity.[13]Most people with COPD die from comorbidities and not from respiratory problems.[48]

Anxiety and depression are often complications of COPD.[2][1]Other complications include a reduced quality of life and increased disability,cor pulmonale,frequent chest infections includingpneumonia,secondary polycythemia,respiratory failure,pneumothorax,lung cancer, andcachexia(muscle wasting).[1][2][49]

Cognitive impairmentis common in those with COPD as it is for other lung conditions that affect airflow. Cognitive impairment is associated with the declining ability to cope with the basicactivities of daily living.[50]

It is unclear if those with COPD are at greater risk of contractingCOVID-19,though if infected they are at risk of hospitalization and developing severe COVID-19. However, there are laboratory and clinical studies showing a possibility of certaininhaled corticosteroidsfor COPD providing a protective role against COVID-19.[51] Differentiating COVID-19 symptoms from an exacerbation is difficult; mildprodromal symptomsmay delay its recognition and where they include loss of taste or smell COVID-19 is to be suspected.[37]

Definition

[edit]

Many definitions of COPD in the past included chronic bronchitis andemphysemabut these have never been included inGOLD reportdefinitions.[8]Emphysema is defined as enlarged airspaces (alveoli) whose walls break down resulting in permanent damage to thelung tissueand is just one of the structural abnormalities that can limit airflow. The condition can exist without airflow limitation but commonly it does.[11]Chronic bronchitis is defined as aproductive coughthat is present for at least three months each year for two years but does not always result in airflow limitation although the risk of developing COPD is great.[13]These older definitions grouped the two types astype Aandtype B.Type A were emphysema types known aspink puffersdue to their pink complexion, fast breathing rate and pursed lips. Type B were chronic bronchitic types referred to asblue bloatersdue to low oxygen levels causing abluish color to the skin and lipsand swollen ankles.[52]These differences were suggested to be due to the presence or not ofcollateral ventilation,evident in emphysema and lacking in chronic bronchitis.[53]This terminology was no longer accepted as useful, as most people with COPD have a combination of both emphysema and airway disease.[52]These are now recognized as the two major phenotypes of COPD — emphysematous phenotype and chronic bronchitic phenotype.[10]

Subtypes

[edit]

It has since been recognized that COPD is more complex, with a diverse group of disorders of differing risk factors and clinical courses that has resulted in a number of subtypes or phenotypes of COPD being accepted and proposed.[54][55]The two classic emphysematous and chronic bronchiticphenotypesare fundamentally different conditions with unique underlying mechanisms.[10]Another subtype of COPD, categorized by some as a separate clinical entity, isasthma-COPD overlap,which is a condition sharing clinical features of bothasthmaand COPD.[56][57]Spirometry measures are inadequate for defining phenotypes and chest X-ray, CT and MRI scans have been mostly employed. Most cases of COPD are diagnosed at a late stage and the use of imaging methods would allow earlier detection and treatment.[10]

The identification and recognition of different phenotypes can guide appropriate treatment approaches. For example, thePDE4 inhibitorroflumilastis targeted at the chronic-bronchitic phenotype.[58]

Two inflammatory phenotypes show a phenotype stability: theneutrophilicinflammatory phenotype and theeosinophilicinflammatory phenotype.[59]Mepolizumab,amonoclonal antibody,has been shown to have benefit in treating the eosinophilic inflammatory type rather than the use of oral corticosteroids, but further studies have been called for.[60]

Another recognized phenotype is the frequent exacerbator.[61]The frequent exacerbator has two or more exacerbations a year, has a poor prognosis and is described as a moderately stable phenotype.[32]

A pulmonary vascular COPD phenotype has been described due to cardiovascular dysfunction.[62]A molecular phenotype ofCFTRdysfunction is shared withcystic fibrosis.[14]A combined phenotype of chronic bronchitis andbronchiectasishas been described with a difficulty noted of determining the best treatment.[63]

The onlygenotypeis theAlpha -1 antitrypsin deficiency(AATD) genetic subtype and this has a specific treatment.[64]

Cause

[edit]

The cause of the development of COPD is the exposure to harmfulparticlesor gases, includingtobacco smoke,that irritate the lung causing inflammation that interacts with a number of host factors. Such exposure needs to be significant or long-term.[8]The greatest risk factor for the development of COPD is tobacco smoke.[17]However, less than 50 percent of heavy smokers develop COPD, so other factors need to be considered, including exposure to indoor and outdoor pollutants, allergens, occupational exposure, and host factors.[31][13]One of the known causes of COPD is the exposure toconstruction dust.The three main types of construction dust aresilica dust,non-silica dust (e.g., dust from gypsum, cement, limestone, marble and dolomite) andwood dust.[65]Host factors include a genetic susceptibility,factors associated with poverty,aging and physical inactivity.Asthmaandtuberculosisare also recognized as risk factors, as the comorbidity of COPD is reported to be 12 times higher in patients with asthma after adjusting for smoking history.[13]In Europeairway hyperresponsivenessis rated as the second most important risk factor after smoking.[13]

A host factor of anairway branching variation,arising duringdevelopmenthas been described.[66]Therespiratory treeis a filter for harmful substances and any variant has the potential to disrupt this. A variation has been found to be associated with the development of chronic bronchitis and another with the development of emphysema. A branch variant in the central airway is specifically associated with an increased susceptibility for the later development of COPD. A genetic association for the variants has been sometimes found withFGF10.[66][67]

Alcohol abusecan lead toalcoholic lung diseaseand is seen to be an independent risk factor for COPD.[68][69] Mucociliary clearanceis disrupted by chronic exposure to alcohol;macrophageactivity is diminished and an inflammatory response promoted.[70][69]The damage leads to a susceptibility for infection, includingCOVID-19,[71]more so when combined with smoking; smoking induces the upregulation of the expression ofACE2,a receptor for theSARS-CoV-2virus.[68]

Smoking

[edit]

The primary risk factor for COPD globally istobacco smokingwith an increased rate of developing COPD shown in smokers and ex-smokers.[8][17]Of those who smoke, about 20% will get COPD,[72]increasing to less than 50% in heavy smokers.[8]In the United States and United Kingdom, of those with COPD, 80–95% are either current or previous smokers.[72][73][74]Several studies indicate that women are more susceptible than men to the harmful effects of tobacco smoke.[75]For the same amount of cigarette smoking, women have a higher risk of COPD than men.[76]In non-smokers, exposure tosecond-hand smoke(passive smoking) is the cause of 1.2 million deaths from the more than 8 million deaths worldwide each year due totobacco smoke.[77]Women who smoke duringpregnancy,and during the early life of the child is a risk factor for the later development of COPD in their child.[78]

Inhaled smoke triggers the release of excessiveproteasesin lungs, which then degradeselastin,the major component of alveoli.[17]Smoke also impairs the action ofcilia,inhibitingmucociliary clearancethat clears the bronchi of mucus, cellular debris and unwanted fluid.[17]

Other types of tobacco smoke, such as from cigar,pipe,water-pipeandhookahuse, also confer a risk.[13]Water-pipe or hookah smoke appears to be as harmful or even more harmful than smoking cigarettes.[79]

Marijuanais the second most commonly smoked substance, but evidence linking its use to COPD is very limited. Limited evidence shows that marijuana does not accelerate lung function decline.[80]A low use of marijuana gives a bronchodilatory effect rather than the bronchoconstrictive effect from tobacco use, but it is often smoked in combination with tobacco or on its own by tobacco smokers. Higher use however has shown a decline in theFEV1.[81]There is evidence of it causing some respiratory problems and its use in combination may have a cumulative toxic effect suggesting it as a risk factor for spontaneous pneumothorax,bullous emphysema,COPD and lung cancer.[80][82]A noted difference between marijuana use and tobacco was that respiratory problems were resolved with stopping usage unlike the continued decline with stopping tobacco smoking.[80]Respiratory symptoms reported with marijuana use included chronic cough, increased sputum production and wheezing but not shortness of breath. Also these symptoms were typically reported ten years ahead of their affecting tobacco smokers.[80]Another study found that chronic marijuana smokers even with the additional use of tobacco developed similar respiratory problems, but did not seem to develop airflow limitation and COPD.[83]

Pollution

[edit]
Access toclean fuelandclean cooking facilitiesas of 2016[84]

Exposure toparticulatescan bring about the development of COPD, or its exacerbations. Those with COPD are more susceptible to the harmful effects of particulate exposure that can cause acute exacerbations brought about by infections.[47]Black carbonalso known assoot,is anair pollutantassociated with an increased risk of hospitalization due to the exacerbations caused. Long-term exposure is indicated as an increased rate of mortality in COPD.[47]Studies have shown that people who live in large cities have a higher rate of COPD compared to people who live in rural areas.[85]Areas with poor outdoor air quality, including that fromexhaust gas,generally have higher rates of COPD.[86]Urban air pollution significantly effects the developing lung and its maturation, and contributes a potential risk factor for the later development of COPD. The overall effect in relation to smoking is believed to be small.[13]

Poorly ventilated fires used for cooking and heating, are often fueled by coal orbiomasssuch as wood anddry dung,leading toindoor air pollutionand are one of the most common causes of COPD indeveloping countries.Women are affected more as they have a greater exposure.[13]These fuels are used as the main source of energy in 80% of homes inIndia,Chinaandsub-Saharan Africa.[86]

Occupational exposure

[edit]

Intense and prolonged exposure to workplacedusts,chemicals and fumes increases the risk of COPD in smokers, nonsmokers and never-smokers. Substances implicated in occupational exposure and listed in the UK, includeorganicandinorganicdusts such ascadmium,silica,dust fromgrainsandflourand fumes from cadmium andweldingthat promote respiratory symptoms.[18][13]Workplace exposure is believed to be the cause in 10–20% of cases and in the United States, it is believed to be related to around 30% of cases among never smokers and probably represents a greater risk in countries without sufficient regulations.[13][87]The negative effects of dust exposure and cigarette smoke exposure appear to be cumulative.[88]

Genetics

[edit]

Genetics play a role in the development of COPD. It is more common among relatives of those with COPD who smoke than unrelated smokers.[13]The most well known genetic risk factor isAlpha -1 antitrypsin deficiency(AATD) and this is the onlygenotype(genetic subtype) with a specific treatment.[64]This risk is particularly high if someone deficient inAlpha -1 antitrypsin(AAT) also smokes.[89]It is responsible for about 1–5% of cases[89][90]and the condition is present in about three to four in 10,000 people.[91]

Mutations inMMP1genethat encodes forinterstitial collagenaseare associated with COPD.[92]

The COPDGene study is an ongoing longitudinal study into the epidemiology of COPD, identifying phenotypes and looking for their likely association with susceptible genes.Genome wide analysesin concert with theInternational COPD Genetics Consortiumhas identified more than 80 genome regions associated with COPD and further studies in these regions has been called for.Whole genome sequencingis an ongoing collaboration (2019) with theNational Heart, Lung and Blood Institute(NHLBI) to identify rare genetic determinants.[93]

Pathophysiology

[edit]
Normallungsshown in upper diagram. Lungs damaged by COPD in lower diagram with an inset showing a cross-section ofbronchiolesblocked bymucusand damagedalveoli.

COPD is a progressivelung diseasein which chronic, incompletely reversible poor airflow (airflow limitation) and an inability to breathe out fully (air trapping) exist.[94]The poor airflow is the result of small airways disease and emphysema (the breakdown oflung tissue).[95]The relative contributions of these two factors vary between people.[8]Air trapping precedes lung hyperinflation.[96]

COPD develops as a significant and chronic inflammatory response to inhaled irritants which ultimately leads to bronchial and alveolar remodelling in the lung known assmall airways disease.[97][98][99]Thus, airway remodelling with narrowing of peripheral airway and emphysema are responsible for the alteration of lung function.[59]Mucociliary clearanceis particularly altered with a dysregulation ofciliaandmucusproduction.[100]Small airway disease sometimes calledchronic bronchiolitis,appears to be the precursor for the development of emphysema.[101] The inflammatory cells involved includeneutrophilsandmacrophages,two types of white blood cells. Those who smoke additionally havecytotoxic T cellinvolvement and some people with COPD haveeosinophilinvolvement similar to that in asthma. Part of this cell response is brought on by inflammatory mediators such aschemotactic factors.Other processes involved with lung damage includeoxidative stressproduced by high concentrations offree radicalsin tobacco smoke and released by inflammatory cells and breakdown of theconnective tissueof the lungs byproteases(particularlyelastase) that are insufficiently inhibited byprotease inhibitors.The destruction of the connective tissue of the lungs leads to emphysema, which then contributes to the poor airflow and finally, poor absorption and release of respiratory gases. General muscle wasting that often occurs in COPD may be partly due to inflammatory mediators released by the lungs into the blood.[13]

Micrographshowingemphysema(left – large empty spaces) andlung tissuewith relative preservedalveoli(right)

Narrowing of the airways occurs due to inflammation and subsequent scarring within them. This contributes to the inability to breathe out fully. The greatest reduction in air flow occurs when breathing out, as the pressure in the chest is compressing the airways at this time.[102]This can result in more air from the previous breath remaining within the lungs when the next breath is started, resulting in an increase in the total volume of air in the lungs at any given time, a process called air trapping which is closely followed byhyperinflation.[102][103][96]Hyperinflation from exercise is linked to shortness of breath in COPD, as breathing in is less comfortable when the lungs are already partly filled.[104]Hyperinflation may also worsen during an exacerbation.[105]There may also be a degree ofairway hyperresponsivenessto irritants similar to those found in asthma.[91]

Low oxygen levelsand eventually,high carbon dioxide levels in the blood,can occur from poorgas exchangedue to decreased ventilation from airway obstruction, hyperinflation and a reduced desire to breathe.[13]During exacerbations, airway inflammation is also increased, resulting in increased hyperinflation, reduced expiratory airflow and worsening of gas transfer. This can lead to low blood oxygen levels which if present for a prolonged period, can result innarrowing of the arteriesin the lungs, while emphysema leads to the breakdown of capillaries in the lungs. Both of these conditions may result inpulmonary heart diseasealso classically known ascor pulmonale.[49]

Diagnosis

[edit]
A person sitting and blowing into a device attached to a computer
A person blowing into aspirometer.Smaller handheld devices are available for office use.

The diagnosis of COPD should be considered in anyone over the age of 35 to 40 who hasshortness of breath,a chronic cough, sputum production, or frequent winter colds and a history of exposure to risk factors for the disease.Spirometryis then used to confirm the diagnosis.[4][106]

Spirometry

[edit]

Spirometrymeasures the amount of airflow obstruction present and is generally carried out after the use of abronchodilator,a medication to open up the airways.[107]Two main components are measured to make the diagnosis, theforced expiratory volume in one second(FEV1), which is the greatest volume of air that can be breathed out in the first second of a breath and theforced vital capacity(FVC), which is the greatest volume of air that can be breathed out in a single large breath.[108]Normally, 75–80% of the FVC comes out in the first second[108]and aFEV1/FVC ratioless than 70% in someone with symptoms of COPD defines a person as having the disease.[107]Based on these measurements, spirometry would lead to over-diagnosis of COPD in the elderly.[107]TheNational Institute for Health and Care Excellencecriteria additionally require a FEV1 less than 80% of predicted.[109]People with COPD also exhibit a decrease indiffusing capacity of the lung for carbon monoxidedue to decreased surface area in the alveoli, as well as damage to the capillary bed.[110]Testing thepeak expiratory flow(the maximum speed of expiration), commonly used in asthma diagnosis, is not sufficient for the diagnosis of COPD.[109]

Screeningusing spirometry in those without symptoms has uncertain effect and is generally not recommended; however, it is recommended for those without symptoms but with a known risk factor.[44]

Assessment

[edit]
MRCshortness of breath scale[109]
Grade Activity affected
1 Only strenuous activity
2 Vigorous walking
3 With normal walking
4 After a few minutes of walking
5 With changing clothing
GOLDcriteria[4]
Severity FEV1% predicted
Mild (GOLD 1) ≥80
Moderate (GOLD 2) 50–79
Severe (GOLD 3) 30–49
Very severe (GOLD 4) <30

A number of methods can be used to assess the affects and severity of COPD.[106][44]TheMRC breathlessness scaleor the COPD assessment test (CAT) are simple questionnaires that may be used.[111][106]GOLDrefers to a modified MRC scale that if used, needs to include other tests since it is simply a test of breathlessness experienced.[44][112]Scores on CAT range from 0–40 with the higher the score, the more severe the disease.[113]Spirometry may help to determine the severity of airflow limitation.[4]This is typically based on the FEV1 expressed as a percentage of the predicted "normal" for the person's age, gender, height and weight.[4]Guidelines published in 2011 by American and European medical societies recommend partly basing treatment recommendations on the FEV1.[107]TheGOLDguidelines group people into four categories based on symptoms assessment, degree of airflow limitation and history of exacerbations.[112]Weight loss, muscle loss and fatigue are seen in severe and very severe cases.[44]

Use of screening questionnaires, such as COPD diagnostic questionnaire (CDQ), alone or in combination with hand-held flow meters is appropriate for screening of COPD in primary care.[114]

Other tests

[edit]

Achest X-rayis not useful to establish a diagnosis of COPD but it is of use in either excluding other conditions or including comorbidities such aspulmonary fibrosisandbronchiectasis.Characteristic signs of COPD on X-ray include hyperinflation (shown by a flattened diaphragm and an increased retrosternal air space) and lung hyperlucency.[5]Asaber-sheath tracheamay also be shown that isindicativeof COPD.[115]

ACT scanis not routinely used except for the exclusion of bronchiectasis.[5]Ananalysis of arterial bloodis used to determine the need for oxygen supplementation and assess forhigh levels of carbon dioxidein the blood; this is recommended in those with an FEV1 less than 35% predicted, those with a peripheral oxygen saturation less than 92% and those with symptoms of congestive heart failure.[116]WHOrecommends that all those diagnosed with COPD be screened forAlpha -1 antitrypsin deficiency.[44]

Differential diagnosis

[edit]

COPD may need to bedifferentiatedfrom other conditions such ascongestive heart failure,asthma,bronchiectasis,tuberculosis,obliterative bronchiolitisanddiffuse panbronchiolitis.[5]The distinction between asthma and COPD is made on the basis of the symptoms, smoking history and whether airflow limitation is reversible with bronchodilators at spirometry.[117]Chronic bronchitis with normal airflow is not classified as COPD.[91]

Prevention

[edit]

Most cases of COPD are potentially preventable through decreasing exposure to tobacco smoke and other indoor and outdoor pollutants.[20]

Smoking cessation

[edit]

Thepoliciesof governments, public health agencies and antismoking organizations can reduce smoking rates by discouraging people from starting and encouraging people to stop smoking.[118]Smoking bansin public areas and places of work are important measures to decrease exposure to secondhand smoke and while many places have instituted bans, more are recommended.[86]

In those who smoke,stopping smokingis the only measure shown to slow down the worsening of COPD.[119][120]Even at a late stage of the disease, it can reduce the rate of worsening lung function and delay the onset of disability and death.[121]Often, several attempts are required before long-term abstinence is achieved.[118]Attempts over 5 years lead to success in nearly 40% of people.[122]

Some smokers can achieve long-term smoking cessation through willpower alone. Smoking, however, is highly addictive and many smokers need further support.[123]The chance of quitting is improved with social support, engagement in a smoking cessation program and the use of medications such asnicotine replacement therapy,bupropion,orvarenicline.[118][120][122]Combining smoking-cessation medication with behavioral therapy is more than twice as likely to be effective in helping people with COPD stop smoking, compared with behavioral therapy alone.[124]

Occupational health

[edit]

A number of measures have been taken to reduce the likelihood that workers in at-risk industries—such as coal mining, construction and stonemasonry—will develop COPD.[86]Examples of these measures include the creation of public policy,[86]education of workers and management about the risks, promoting smoking cessation,checkingworkers for early signs of COPD, use ofrespiratorsand dust control.[125][126]Effective dust control can be achieved by improving ventilation, using water sprays and by using mining techniques that minimize dust generation.[127]If a worker develops COPD, further lung damage can be reduced by avoiding ongoing dust exposure, for example by changing their work role.[128]

Pollution control

[edit]

Both indoor and outdoor air quality can be improved, which may prevent COPD or slow the worsening of existing disease.[86]This may be achieved by public policy efforts, cultural changes and personal involvement.[129]Many developed countries have successfully improved outdoor air quality through regulations which has resulted in improvements in the lung function of their populations.[86]Individuals are also advised to avoid irritants of indoor and outdoor pollution.[20]

In developing countries one key effort is to reduce exposure to smoke from cooking and heating fuels through improved ventilation of homes and better stoves and chimneys.[129]Proper stoves may improveindoor air qualityby 85%. Using alternative energy sources such assolar cookingand electrical heating is also effective. Using fuels such as kerosene or coal might produce less household particulate matter than traditional biomass such as wood or dung, but whether this is better health wise is unclear.[86]

Management

[edit]

COPD currently has no cure,[130]but the symptoms are treatable and its progression can be delayed, particularly by stopping smoking.[1][6]The major goals of management are to reduce exposure to risk factors including offering non-pharmacological treatments such as help with stopping smoking. Stopping smoking can reduce the rate of lung function decline and also reduce mortality from smoking-related diseases such as lung cancer and cardiovascular disease.[1]Other recommendations include annualinfluenza vaccinationsandpneumococcal vaccinationto help reduce the risk of exacerbations; CDC and GOLD 2024 also recommendsRSVvaccine for individuals above 60 years;[131][132]giving advice as to healthy eating and encouraging physical exercise. Guidance is also advised as to managing breathlessness and stress.[6]

Other illnessesare also managed. An action plan is drawn up and is to be reviewed.[20]Providing people with a personalized action plan, an educational session and support for use of their action plan in the event of an exacerbation, reduces the number of hospital visits and encourages early treatment of exacerbations.[133]When self-management interventions, such as taking corticosteroids and using supplemental oxygen, is combined with action plans, health-related quality of life is improved compared to usual care.[134]In those with COPD who aremalnourished,supplementation withvitamin C,vitamin E,zincandseleniumcan improve weight, strength ofrespiratory musclesand health-related quality of life.[21]Significantvitamin D deficiencyis common in those with COPD and can cause increased exacerbations. Supplementation when deficient can give a 50% reduction in the number of exacerbations.[32][135]

A number ofmedical treatmentsare used in the management of stable COPD and exacerbations. These includebronchodilators,corticosteroidsandantibiotics.

In those with a severe exacerbation, antibiotics improve outcomes.[136]A number of different antibiotics may be used includingamoxicillin,doxycyclineandazithromycin;whether one is better than the others is unclear.[137]There is no clear evidence of improved outcomes for those with less severe cases.[136]The FDA recommends against the use offluoroquinoloneswhen other options are available due to higher risks of serious side effects.[138] In treatingacute hypercapnic respiratory failure(acutely raised levels of carbon dioxide),bilevel positive airway pressure(BPAP) can decrease mortality and the need ofintensive care.[139]Fewer than 20% of exacerbations require hospital admission.[129]In those without acidosis from respiratory failure,home caremay be able to help avoid some admissions.[129]

In those withend-stage disease,palliative careis focused on relieving symptoms.[140]Morphinecan improve exercise tolerance.[21]Non-invasive ventilationmay be used to support breathing and also reduce daytime breathlessness.[141][21]

Bronchodilators

[edit]

Inhaled short-actingbronchodilatorsare the primary medications used on anas neededbasis; their use on a regular basis is not recommended.[6]The two major types arebeta2-adrenergic agonistsandanticholinergics;either in long-acting or short-acting forms. Beta2–adrenergic agonists targetreceptorsin thesmooth muscle cellsinbronchiolescausing them to relax and allow improved airflow. They reduce shortness of breath, tend to reducedynamic hyperinflationand improve exercise tolerance.[6][142]Short-acting bronchodilators have an effect for four hours and for maintenance therapy long acting bronchodilators with an effect of over twelve hours are used. In times of more severe symptoms a short acting agent may be used in combination.[6]An inhaled corticosteroid used with a long-acting beta-2 agonist is more effective than either one on its own.[143]

Which type of long-acting agent,long-acting muscarinic antagonist(LAMA) such astiotropiumorlong-acting beta agonist(LABA), is better is unclear and trying each and continuing with the one that works best may be advisable.[144]Both types of agent appear to reduce the risk of acute exacerbations by 15–25%.[139]The combination of LABA/LAMA may reduce COPD exacerbations and improve quality-of-life compared to long-acting bronchodilators alone.[145]The 2018 NICE guideline recommends use of dual long-acting bronchodilators with economic modelling suggesting that this approach is preferable to starting one long acting bronchodilator and adding another later.[146]

Several short-acting β2agonists are available, includingsalbutamol(albuterol) andterbutaline.[129]They provide some relief of symptoms for four to six hours.[129]Along-acting beta agonist(LABA) such assalmeterol,formoterolandindacaterolare often used as maintenance therapy. Some feel the evidence of benefits is limited,[147]while others view the evidence of benefit as established.[148][149][150]Long-term use appears safe in COPD[151]with adverse effects includeshakinessandheart palpitations.[139]When used with inhaled steroids they increase the risk of pneumonia.[139]While steroids and LABAs may work better together,[147]it is unclear if this slight benefit outweighs the increased risks.[152]There is some evidence that combined treatment of LABAs with long-actingmuscarinic antagonists(LAMA), an anticholinergic, and LABA +ICS (inhaled corticosteroid) may be similar in benefits in terms of fewer exacerbation's and quality of life measures for moderate to severe COPD, but LAMA+LABA offers better improvements in forced expiratory volume (FEV1%) and a lower risk of pneumonia.[153]All three together, LABA, LAMA and ICS, have some evidence of benefits.[154]Indacaterol requires an inhaled dose once a day and is as effective as the other long-acting β2agonist drugs that require twice-daily dosing for people with stable COPD.[150]

The two main anticholinergics used in COPD areipratropiumandtiotropium.Ipratropium is a short-acting muscarinic antagonist (SAMA), while tiotropium is long-acting (LAMA). Tiotropium is associated with a decrease in exacerbations and improved quality of life,[155]and tiotropium provides those benefits better than ipratropium.[156]It does not appear to affect mortality or the overall hospitalization rate.[155]Anticholinergics can cause dry mouth and urinary tract symptoms.[139]They are also associated with increased risk of heart disease andstroke.[157]Aclidinium,another long-acting agent, reduces hospitalizations associated with COPD and improves quality of life.[158][159][160]The LAMAumeclidinium bromideis another anticholinergic alternative.[161]When compared to tiotropium, the LAMAs aclidinium, glycopyrronium, and umeclidinium appear to have a similar level of efficacy; with all four being more effective thanplacebo.[162]Further research is needed comparing aclidinium to tiotropium.[160]

Corticosteroids

[edit]

Inhaledcorticosteroidsare anti-inflammatories that are recommended byGOLDas a first-line maintenance treatment in COPD cases with repeated exacerbations.[163][164]Their regular use increases the risk of pneumonia in severe cases.[32]Studies have shown that the risk of pneumonia is associated with all types of corticosteroids; is related to the disease severity and a dose-response relationship has been noted.[163]Oralglucocorticoidscan be effective in treating an acute exacerbation.[143]They appear to have fewer side effects than those given intravenously.[165]Five days of steroids work as well as ten or fourteen days.[166]

The use of corticosteroids is associated with a decrease in the number of lymphoid follicles (in thebronchial lymphoid tissue).[101]A triple inhaled therapy of LABA/LAMA/ICS improves lung function, reduces symptoms and exacerbations and is seen to be more effective than mono or dual therapies.[167][143]NICE guidelines recommend the use of ICSs in people with asthmatic features or features suggesting steroid responsiveness.[146]

PDE4 inhibitors

[edit]

Phosphodiesterase-4 inhibitors(PDE4 inhibitors) are anti-inflammatories that improve lung function and reduce exacerbations in moderate to severe illness.Roflumilastis a PDE4 inhibitor used orally once daily to reduce inflammation, it has no direct bronchodilatory effects. It is essentially used in treating those with chronic bronchitis along with systemic corticosteroids.[60]Reported adverse effects of roflumilast appear early in treatment, become less with continued treatment and are reversible. One effect is dramatic weight loss and its use is to be avoided in underweight people. It is also advised to be used with caution in those who have depression.[60]

Other medications

[edit]

Long-termpreventive useofantibiotics,specifically those from themacrolideclass such aserythromycin,reduce the frequency of exacerbations in those who have two or more a year.[168][169]This practice may be cost effective in some areas of the world.[170]Concerns include the potential forantibiotic resistanceand side effects includinghearing loss,tinnitusand changes to the heart rhythm known aslong QT syndrome.[169]

Methylxanthinessuch astheophyllineare widely used. Theophylline is seen to have a mild bronchodilatory effect in stable COPD. Inspiratory muscle function is seen to be improved but the causal effect is unclear. Theophylline is seen to improve breathlessness when used as an add-on tosalmeterol.All instances of improvement have been reported using sustained release preparations.[6]Methylxanthines are not recommended for use in exacerbations due to adverse effects.[32]

Mucolyticsmay help to reduce exacerbations in some people with chronic bronchitis; noticed by less hospitalization and less days of disability in one month.[171]Erdosteineis recommended by NICE.[172]GOLD also supports the use of some mucolytics that are advised against when inhaled corticosteroids are being used and singles out erdosteine as having good effects regardless of corticosteroid use. Erdosteine also has antioxidant properties but there is not enough evidence to support the general use of antioxidants.[60]Erdosteine has been shown to significantly reduce the risk of exacerbations, shorten their duration and hospital stays.[173]

Cough medicinesare not recommended.[174]Beta blockersare not contraindicated for those with COPD and should only be used where there is concomitant cardiovascular disease.[60]

Recent studies show that metformin plays a role in reducing systemic inflammation by reducing biomarker levels that are increased during COPD exacerbations.[175]

Oxygen therapy

[edit]

Supplemental oxygenis recommended for those withlow oxygen levelsinrespiratory failureat rest (apartial pressure of oxygenless than 50–55 mmHg oroxygen saturationsof less than 88%).[21]When taking into account complications includingcor pulmonaleand pulmonary hypertension, the levels involved are 56–59 mmHg.[176]Oxygen therapy is to be used for between 15 and 18 hours per day and is said to decrease the risk ofheart failureand death.[176]In those with normal or mildly low oxygen levels, oxygen supplementation (ambulatory) may improve shortness of breath when given during exercise, but may not improve breathlessness during normal daily activities or affect the quality of life.[177]During acute exacerbations, many require oxygen therapy; the use of high concentrations of oxygen without taking into account a person's oxygen saturations may lead to increased levels of carbon dioxide and worsened outcomes.[178][179]In those at high risk of high carbon dioxide levels, oxygen saturations of 88–92% are recommended, while for those without this risk, recommended levels are 94–98%.[179]Once prescribed long-term oxygen therapy, patients should be re-assessed after 60 to 90 days, to deteermine whether supplemental oxygen is still indicated and if prescribed supplemental oxygen is effective.[180][181]

Rehabilitation

[edit]

Pulmonary rehabilitationis a program of exercise, disease management and counseling, coordinated to benefit the individual.[182]A severe exacerbation leads to hospital admission, high mortality and a decline in the ability to carry out daily activities. Following a hospital admission pulmonary rehabilitation has been shown to significantly reduce future hospital admissions, mortality and improve quality of life.[58]

The optimal exercise routine, use of noninvasive ventilation during exercise and intensity of exercise suggested for people with COPD, is unknown.[183][184]Performing endurance arm exercises improves arm movement for people with COPD and may result in a small improvement in breathlessness.[185]Performing arm exercises alone does not appear to improve quality of life.[185]Pursed-lip breathingexercises may be useful.[28]Tai chiexercises appear to be safe to practice for people with COPD and may be beneficial for pulmonary function and pulmonary capacity when compared to a regular treatment program.[186]Tai Chi was not found to be more effective than other exercise intervention programs.[186]Inspiratory and expiratory muscle training (IMT, EMT) have been suggested and may provide some improvements when compared to no treatment.[187]A combination of IMT and walking exercises at home may help limit breathlessness in cases of severe COPD.[188]Additionally, the use of low amplitude high velocity joint mobilization together with exercise improves lung function and exercise capacity.[189]The goal of spinal manipulation therapy is to improve thoracic mobility in an effort to reduce the work on the lungs during respiration, however, the evidence supporting manual therapy for people with COPD is very weak.[189][190]

Airway clearance techniques(ACTs), such aspostural drainage,percussion/vibration,autogenic drainage,hand-heldpositive expiratory pressure(PEP) devices and other mechanical devices, may reduce the need for increased ventilatory assistance, the duration of ventilatory assistance and the length of hospital stay in people with acute COPD.[191]In people with stable COPD, ACTs may lead to short-term improvements in health-related quality of life and a reduced long-term need for hospitalizations related to respiratory issues.[191]

Being either underweight or overweight can affect the symptoms, degree of disability and prognosis of COPD. People with COPD who are underweight can improve their breathing muscle strength by increasing their calorie intake. When combined with regular exercise or a pulmonary rehabilitation program, this can lead to improvements in COPD symptoms. Supplemental nutrition may be useful in those who aremalnourished.[21][192]

Management of exacerbations

[edit]

People with COPD can experienceexacerbations(flare-ups) that are commonly caused byrespiratory tractinfections. The symptoms that worsen are not specific to COPD and differential diagnoses need to be considered.[32]Acute exacerbations are typically treated by increasing the use of short-acting bronchodilators including a combination of a short-acting inhaled beta agonist and short-acting anticholinergic.[32]These medications can be given either via ametered-dose inhalerwith aspaceror via anebulizer,with both appearing to be equally effective.[129][193]Nebulization may be easier for those who are more unwell.[129]Oxygen supplementationcan be useful. Excessive oxygen; however, can result inincreased CO2levelsand a decreased level of consciousness.[194]Corticosteroidsgiven orallycan improve lung function and shorten hospital stays but their use is recommended for only five to seven days; longer courses increase the risk of pneumonia and death.[32]

Room temperatures

[edit]

Maintainingroom temperaturesof at least 21 °C (70 °F) for a minimum of nine hours a day was associated with better health in those with COPD, especially for smokers.[195]The World Health Organization (WHO) recommends indoor temperatures of a slightly higher range between 18 and 24 °C (64 and 75 °F).[196]

Room humidity

[edit]

For people with COPD, the ideal indoor humidity levels are 30–50% RH. Maintaining indoor humidity can be difficult in the winter, especially in cold climates where the heating system is constantly running.[197]

Keeping the indoor relative humidity above 40% RH significantly reduces the infectivity of aerosolized viruses.[198]

Procedures for emphysema

[edit]

There are a number of procedures toreduce the volume of a lungin cases of severe emphysema with hyperinflation.

Surgical

[edit]

For severe emphysema that has proved unresponsive to other therapieslung volume reduction surgery(LVRS) may be an option.[199][200]LVRS involves the removal of damaged tissue, which improves lung function by allowing the rest of the lungs to expand.[139][129]It is considered when the emphysema is in the upper lobes and when there are no comorbidities.[201]

Bronchoscopic

[edit]

Minimally invasive bronchoscopic procedures may be carried out to reduce lung volume. These include the use of valves, coils, or thermal ablation.[21][202]Endobronchial valvesare one-way valves that may be used in those with severe hyperinflation resulting from advanced emphysema; a suitable target lobe and nocollateral ventilationare required for this procedure. The placement of one or more valves in the lobe induces a partialcollapseof the lobe that ensures a reduction in residual volume that improves lung function, the capacity for exercise and quality of life.[203]

The placement ofnitinolcoils instead of valves is recommended where there is collateral ventilation that would prevent the use of valves.[204]Nitinol is abiocompatible alloy.

Both of these techniques are associated with adverse effects including persistent air leaks and cardiovascular complications. Thermal vapor ablation has an improved profile. Heated water vapor is used to target lobe regions which leads to permanent fibrosis and volume reduction. The procedure is able to target individual lobe segments, can be carried out regardless of collateral ventilation and can be repeated with the natural advance of emphysema.[205]

Other surgeries

[edit]

In very severe caseslung transplantationmight be considered.[199]A CT scan may be useful in surgery considerations.[91]Ventilation/perfusion scintigraphyis another imaging method that may be used to evaluate cases for surgical interventions and also to evaluate post-surgery responses.[206]Abullectomymay be carried out when a giantbullaoccupies more than a third of a hemithorax.[201]

Prognosis

[edit]
Chronic obstructive pulmonary disease deaths per million persons in 2012:
9–63
64–80
81–95
96–116
117–152
153–189
190–235
236–290
291–375
376–1089
Disability-adjusted life yearslost to chronic obstructive pulmonary disease per 100,000 inhabitants in 2004:[207]

COPD is progressive and can lead to premature death. It is estimated that 3% ofall disabilityis related to COPD.[208]The proportion of disability from COPD globally has decreased from 1990 to 2010 due to improved indoor air quality primarily in Asia.[208]The overall number of years lived with disability from COPD, however, has increased.[209]

There are many variables affecting the long-term outcome in COPD and GOLD recommends the use of a composite test (BODE) that includes the main variables ofbody-mass index,obstruction of airways,dyspnea(breathlessness) and exercise and not just spirometry results.[210]

NICE recommends against the use of BODE for the prognosis assessment in stable COPD; factors such as exacerbations andfrailtyneed to be considered.[202]Other factors that contribute to a poor outcome include older age,comorbiditiessuch as lung cancer and cardiovascular disease and the number and severity of exacerbations needing hospital admittance.[32]

Epidemiology

[edit]

Estimates of prevalence have considerable variation due to differences in analytical and surveying approach and the choice of diagnostic criteria.[211]An estimated 384 million people aged 30 years or more had COPD in 2010, corresponding to a global prevalence of 12%.[8]The disease affects men and women.[3]The increase in the developing world between 1970 and the 2000s is believed to be related to increasing rates of smoking in this region, an increasing population and an aging population due to fewer deaths from other causes such as infectious diseases.[139]Some developed countries have seen increased rates, some have remained stable and some have seen a decrease in COPD prevalence.[139]

Around three million people die of COPD each year.[8]In some countries, mortality has decreased in men but increased in women.[212]This is most likely due to rates of smoking in women and men becoming more similar.[91]A higher rate of COPD is found in those over 40 years and this increases greatly with advancing age with the highest rate found in those over 60 years.[8]Sex differences in the anatomy of the respiratory system include smaller airway lumens and thicker airway walls in women, which contribute to a greater severity of COPD symptoms like dyspnea and frequency of COPD exacerbation.[213]

In the UK, three million people are reported to be affected by COPD – two million of these being undiagnosed. On average, the number of COPD-related deaths between 2007 and 2016 was 28,600. The estimated number of deaths due to occupational exposure was estimated to be about 15% at around 4,000.[211]In the United States in 2018, almost 15.7 million people had been diagnosed with COPD and it is estimated that millions more have not been diagnosed.[214]

In 2011, there were approximately 730,000 hospitalizations in the United States for COPD.[215]Globally, COPD in 2019 was the third-leading cause of death. In low-income countries, COPD does not appear in the Top 10 causes of death; in other income groups, it is in the Top 5.[216]

History

[edit]
Giovanni Battista Morgagni,who made one of the earliest recorded descriptions of emphysema in 1769

The namechronic obstructive pulmonary diseaseis believed to have first been used in 1965.[217]Previously it has been known by a number of different names, includingchronic obstructive bronchopulmonary disease,chronic airflow obstruction,chronic obstructive lung disease,nonspecific chronic pulmonary disease,diffuse obstructive pulmonary syndrome.[217]

The termsemphysemaandchronic bronchitiswere formally defined as components of COPD in 1959 at theCIBAguest symposium and in 1962 at theAmerican Thoracic SocietyCommittee meeting on Diagnostic Standards.[217]

Early descriptions of probable emphysema began in 1679 by T. Bonet of a condition of "voluminous lungs" and in 1769 byGiovanni Morgagniof lungs which were "turgid particularly from air".[217][218]In 1721 the first drawings of emphysema were made by Ruysh.[218]René Laennec,used the termemphysemain his bookA Treatise on the Diseases of the Chest and of Mediate Auscultation(1837) to describe lungs that did not collapse when he opened the chest during an autopsy. He noted that they did not collapse as usual because they were full of air and the airways were filled with mucus.[217]In 1842,John Hutchinsoninvented thespirometer,which allowed the measurement ofvital capacityof the lungs. However, his spirometer could only measure volume, not airflow. Tiffeneau and Pinelli in 1947 described the principles of measuring airflow.[217]

Air pollution and the increase in cigarette smoking in Great Britain at the start of the 20th century led to high rates of chronic lung disease, though it received little attention until theGreat Smog of Londonin December 1952. This spurred epidemiological research in the United Kingdom, Holland and elsewhere.[219]In 1953, George L. Waldbott, an American allergist, first described a new disease he namedsmoker's respiratory syndromein the 1953Journal of the American Medical Association.This was the first association between tobacco smoking and chronic respiratory disease.[220]

Modern treatments were developed during the second half of the 20th century. Evidence supporting the use ofsteroidsin COPD was published in the late 1950s.Bronchodilatorscame into use in the 1960s following a promising trial ofisoprenaline.Further bronchodilators, such as short-actingsalbutamol,were developed in the 1970s and the use oflong-acting bronchodilatorsbegan in the mid-1990s.[221]

Society and culture

[edit]

It is generally accepted that COPD is widely underdiagnosed and many people remain untreated. In the US theNIHhas promoted November asCOPD Awareness Monthto be an annual focus on increasing awareness of the condition.[222]

Economics

[edit]

Globally, as of 2010, COPD is estimated to result in economic costs of $2.1 trillion, half of which occurring in the developing world.[223]Of this total an estimated $1.9 trillion are direct costs such as medical care, while $0.2 trillion are indirect costs such as missed work.[224]This is expected to more than double by 2030.[223]In Europe, COPD represents 3% of healthcare spending.[129]In the United States, costs of the disease are estimated at $50 billion, most of which is due to exacerbation.[116]COPD was among the most expensive conditions seen in U.S. hospitals in 2011, with a total cost of about $5.7 billion.[215]

Research

[edit]

Hyaluronanis anatural sugarin theextracellular matrixthat provides a protective coating for cells. It has been shown that on exposure to pollution the hyaluronan in the lungs breaks down into fragments causing irritation and activation of the immune system. There follows subsequent airway constriction and inflammation. The study showed that the inhalation of unfragmented hyaluronan overcame the effects of fragmented HA and reduced inflammation. Inhaled HA only acts locally in thebronchial treeand does not interfere with any drug. It improves mucus clearance by allowing it to move more freely. Further studies are to be carried out in the US to determine optimum dosage levels.[225]

A newcryogenic treatmentaimed at the chronic bronchitic subtype using aliquid nitrogen meteredcryosprayis being trialed and was due to complete in September 2021.[226][227]

Stem-cell therapyusingmesenchymal stem cellshas the potential to restore lung function and thereby improve quality of life. In June 2021 eightclinical trialshad been completed and seventeen were underway. Overall, stem cell therapy has proved to be safe. The trials include the use of stem cells from different sources such asadipose tissue,bone marrowandumbilical cord blood.[228]

A procedure known astargeted lung denervationis being trialed and has been used as part of a clinical trial (2021) in a hospital in the UK. The newminimally invasive procedurewhich takes about an hour to carry out, places electrodes to destroy branches of thevagus nervein the lungs. The vagus nerve is responsible for bothmuscle contractionand mucus secretion, which results in narrowing the airways. In those with COPD these nerves are overactive, usually as a result of smoking damage and the constant mucus secretion and airway constriction leads to the symptoms of cough, shortness of breath, wheeze and tightness of the chest.[229]

The effectiveness ofAlpha -1 antitrypsinaugmentation treatment for people who have Alpha -1 antitrypsin deficiency is unclear.[230]A later clinical trial of double-dosing has shown some improvements in slowing the breakdown of elastin and the progression of emphysema with further studies being called for.[231]

Mass spectrometryis being studied as a diagnostic tool in COPD.[232]

Research continues into the use oftelehealthcareto treat people with COPD when they experience episodes of shortness of breath; treating people remotely may reduce the number of emergency-room visits and improve the person's quality of life.[233]

Evidence is growing for the effectiveness ofAstaxanthinagainst lung disease including COPD. Astaxanthin is a potent antioxidant with anti-inflammatory properties and more trials are said to be needed into its use.[234]

American COPD patients and their caregivers consider the following COPD-related research areas as the most important:

  • family/social/community research
  • patients' well‐being
  • curative research
  • biomedical therapies
  • policy
  • holistic therapies.[235]

Other animals

[edit]

Chronic obstructive pulmonary disease may occur in a number of other animals and may be caused by exposure to tobacco smoke.[236]Most cases of the disease, however, are relatively mild.[237]Inhorsesit is known asrecurrent airway obstruction(RAO) orheaves.RAO can be quite severe and most often is linked to exposure to common allergens.[238]COPD is also commonly found in old dogs.[239]

See also

[edit]

References

[edit]
  1. ^abcdefghi"Chronic obstructive pulmonary disease".nice.org.Retrieved5 July2021.
  2. ^abc"Chronic obstructive pulmonary disease (COPD) — Complications".BMJ Best Practice.Retrieved11 July2021.
  3. ^abcdefg"Chronic obstructive pulmonary disease (COPD)".Fact Sheets.World Health Organization.Retrieved1 July2021.
  4. ^abcdefghijklGold Report 2021,pp. 20–27, Chapter 2: Diagnosis and initial assessment.
  5. ^abcdGold Report 2021,pp. 33–35, Chapter 2: Diagnosis and initial assessment.
  6. ^abcdefghijkGold Report 2021,pp. 40–46, Chapter 3: Evidence supporting prevention and maintenance therapy.
  7. ^abGBD 2015 Disease and Injury Incidence and Prevalence Collaborators (October 2016)."Global, regional, and national incidence, prevalence, and years lived with disability for 310 diseases and injuries, 1990–2015: a systematic analysis for the Global Burden of Disease Study 2015".Lancet.388(10053): 1545–1602.doi:10.1016/S0140-6736(16)31678-6.PMC5055577.PMID27733282.
  8. ^abcdefghijklGold Report 2021,pp. 4–8, Chapter 1: Definition and overview.
  9. ^"2024 GOLD Report".Global Initiative for Chronic Obstructive Lung Disease - GOLD.Retrieved2024-02-23.
  10. ^abcdeMyc LA, Shim YM, Laubach VE, Dimastromatteo J (April 2019)."Role of medical and molecular imaging in COPD".Clin Transl Med.8(1): 12.doi:10.1186/s40169-019-0231-z.PMC6465368.PMID30989390.
  11. ^ab"ICD-11 - ICD-11 for Mortality and Morbidity Statistics".icd.who.int.Retrieved30 June2021.
  12. ^Martini K, Frauenfelder T (November 2020)."Advances in imaging for lung emphysema".Ann Transl Med.8(21): 1467.doi:10.21037/atm.2020.04.44.PMC7723580.PMID33313212.
  13. ^abcdefghijklmnopqrGold Report 2021,pp. 8–14, Chapter 1: Definition and overview.
  14. ^abDe Rose V, Molloy K, Gohy S, Pilette C, Greene CM (2018)."Airway Epithelium Dysfunction in Cystic Fibrosis and COPD".Mediators Inflamm.2018:1309746.doi:10.1155/2018/1309746.PMC5911336.PMID29849481.
  15. ^GINA and GOLD joint guidelines Ga (2014)."Asthma COPD and asthma COPD overlap syndrome (ACOS)"(PDF).GINA Guidelines.
  16. ^Agusti À, Soriano JB (January 2008)."COPD as a Systemic Disease".COPD: Journal of Chronic Obstructive Pulmonary Disease.5(2): 133–138.doi:10.1080/15412550801941349.ISSN1541-2555.PMID18415812.S2CID32732993.
  17. ^abcde"Chronic obstructive pulmonary disease (COPD) - Aetiology | BMJ Best Practice".bestpractice.bmj.Retrieved25 November2022.
  18. ^ab"COPD causes - occupations and substances".hse.gov.uk.Retrieved3 July2021.
  19. ^Torres-Duque CA, García-Rodriguez MC, González-García M (August 2016). "Is Chronic Obstructive Pulmonary Disease Caused by Wood Smoke a Different Phenotype or a Different Entity?".Archivos de Bronconeumologia.52(8): 425–31.doi:10.1016/j.arbres.2016.04.004.PMID27207325.
  20. ^abcdGold Report 2021,pp. 80–83, Chapter 4: Management of stable COPD.
  21. ^abcdefgGold Report 2021,pp. 60–65, Chapter 3: Evidence supporting prevention and maintenance therapy.
  22. ^Dobler CC, Morrow AS, Beuschel B, Farah MH, Majzoub AM, Wilson ME, et al. (March 2020). "Pharmacologic Therapies in Patients With Exacerbation of Chronic Obstructive Pulmonary Disease: A Systematic Review With Meta-analysis".Annals of Internal Medicine.172(6): 413–422.doi:10.7326/M19-3007.PMID32092762.S2CID211476101.
  23. ^Wang H, Naghavi M, Allen C, Barber RM, Bhutta ZA, Carter A, et al. (GBD 2015 Mortality and Causes of Death Collaborators) (October 2016)."Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015".Lancet.388(10053): 1459–1544.doi:10.1016/S0140-6736(16)31012-1.PMC5388903.PMID27733281.
  24. ^GBD 2013 Mortality and Causes of Death Collaborators (January 2015)."Global, regional, and national age-sex specific all-cause and cause-specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013".Lancet.385(9963): 117–71.doi:10.1016/S0140-6736(14)61682-2.PMC4340604.PMID25530442.
  25. ^"The top 10 causes of death".who.int.Retrieved2024-08-12.
  26. ^"COPD Costs".cdc.gov.5 July 2019. Archived fromthe originalon 9 February 2020.
  27. ^"COPD commissioning toolkit"(PDF).assets.publishing.service.gov.uk.Retrieved18 July2021.
  28. ^abMayer AF, Karloh M, Dos Santos K, de Araujo CL, Gulart AA (March 2018). "Effects of acute use of pursed-lips breathing during exercise in patients with COPD: a systematic review and meta-analysis".Physiotherapy.104(1): 9–17.doi:10.1016/j.physio.2017.08.007.PMID28969859.
  29. ^Gold Report 2021,pp. 90–96, Chapter 4: Management of stable COPD.
  30. ^O'Donnell DE, Milne KM, James MD, de Torres JP, Neder JA (January 2020)."Dyspnea in COPD: New Mechanistic Insights and Management Implications".Advances in Therapy.37(1): 41–60.doi:10.1007/s12325-019-01128-9.PMC6979461.PMID31673990.
  31. ^abSzalontai K, Gémes N, Furák J, et al. (June 2021)."Chronic Obstructive Pulmonary Disease: Epidemiology, Biomarkers, and Paving the Way to Lung Cancer".J Clin Med.10(13): 2889.doi:10.3390/jcm10132889.PMC8268950.PMID34209651.
  32. ^abcdefghijGold Report 2021,pp. 104–109, Chapter 5: Management of exacerbations.
  33. ^"Common Colds".Centers for Disease Control and Prevention.7 October 2020. Archived fromthe originalon June 18, 2023.Retrieved20 August2021.
  34. ^Guo-Parke H, Linden D, Weldon S, Kidney JC, Taggart CC (2020)."Mechanisms of Virus-Induced Airway Immunity Dysfunction in the Pathogenesis of COPD Disease, Progression, and Exacerbation".Frontiers in Immunology.11:1205.doi:10.3389/fimmu.2020.01205.PMC7325903.PMID32655557.
  35. ^Short B, Carson S, Devlin AC, et al. (March 2021)."Non-typeable Haemophilus influenzae chronic colonization in chronic obstructive pulmonary disease (COPD)".Critical Reviews in Microbiology.47(2): 192–205.doi:10.1080/1040841X.2020.1863330.PMID33455514.S2CID230608674.
  36. ^US EPA O (19 April 2016)."Particulate Matter (PM) Basics".epa.gov.Retrieved21 July2021.
  37. ^abHalpin DM, Criner GJ, Papi A, Singh D, Anzueto A, Martinez FJ, Agusti AA, Vogelmeier CF (January 2021)."Global Initiative for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease. The 2020 GOLD Science Committee Report on COVID-19 and Chronic Obstructive Pulmonary Disease".Am J Respir Crit Care Med.203(1): 24–36.doi:10.1164/rccm.202009-3533SO.PMC7781116.PMID33146552.
  38. ^Iacobucci G (August 2020)."Covid lockdown: England sees fewer cases of colds, flu, and bronchitis".BMJ.370:m3182.doi:10.1136/bmj.m3182.PMID32784206.S2CID221097739.
  39. ^US EPA O (13 August 2019)."Health Effects Attributed to Wildfire Smoke".epa.gov.Retrieved21 July2021.
  40. ^"Forest Fires and Lung Health".the lung association.25 August 2014.Retrieved21 July2021.
  41. ^"Bushfire smoke".National Centre for Farmer Health.19 March 2014.Retrieved21 July2021.
  42. ^Luo J, Zhang D, Tang W, Dou LY, Sun Y (2021)."Impact of Frailty on the Risk of Exacerbations and All-Cause Mortality in Elderly Patients with Stable Chronic Obstructive Pulmonary Disease".Clin Interv Aging.16:593–601.doi:10.2147/CIA.S303852.PMC8053481.PMID33880018.
  43. ^Aleva FE, Voets LW, Simons SO, de Mast Q, van der Ven AJ, Heijdra YF (March 2017). "Prevalence and Localization of Pulmonary Embolism in Unexplained Acute Exacerbations of COPD: A Systematic Review and Meta-analysis".Chest.151(3): 544–554.doi:10.1016/j.chest.2016.07.034.PMID27522956.S2CID7181799.
  44. ^abcdefGold Report 2021,pp. 26–33, Chapter 2: Diagnosis and initial assessment.
  45. ^Edgar RG, Patel M, Bayliss S, Crossley D, Sapey E, Turner AM (2017)."Treatment of lung disease in Alpha -1 antitrypsin deficiency: a systematic review".Int J Chron Obstruct Pulmon Dis.12:1295–1308.doi:10.2147/COPD.S130440.PMC5422329.PMID28496314.
  46. ^Gold Report 2021,pp. 26–33, 2. Diagnosis and initial assessment.
  47. ^abcdClementi EA, Talusan A, Vaidyanathan S, Veerappan A, Mikhail M, Ostrofsky D, et al. (January 2019)."Metabolic Syndrome and Air Pollution: A Narrative Review of Their Cardiopulmonary Effects".Toxics.7(1): 6.doi:10.3390/toxics7010006.PMC6468691.PMID30704059.
  48. ^Chan SH, Selemidis S, Bozinovski S, Vlahos R (June 2019)."Pathobiological mechanisms underlying metabolic syndrome (MetS) in chronic obstructive pulmonary disease (COPD): clinical significance and therapeutic strategies".Pharmacol Ther.198:160–188.doi:10.1016/j.pharmthera.2019.02.013.PMC7112632.PMID30822464.
  49. ^abForfia PR, Vaidya A, Wiegers SE (January 2013)."Pulmonary heart disease: The heart-lung interaction and its impact on patient phenotypes".Pulm Circ.3(1): 5–19.doi:10.4103/2045-8932.109910.PMC3641739.PMID23662171.
  50. ^Gold Report 2021,pp. 121–126, Chapter 6: COPD and comorbidities.
  51. ^Singh D, Mathioudakis AG, Higham A (March 2022)."Chronic obstructive pulmonary disease and COVID-19: interrelationships".Curr Opin Pulm Med.28(2): 76–83.doi:10.1097/MCP.0000000000000834.PMC8815646.PMID34690257.
  52. ^abWeinberger SE (2019)."6. Chronic Obstructive Pulmonary Disease".Principles of pulmonary medicine(7th ed.). Elsevier. p. 104.ISBN978-0-323-52371-4.
  53. ^Delaunois L (October 1989)."Anatomy and physiology of collateral respiratory pathways".The European Respiratory Journal.2(9): 893–904.doi:10.1183/09031936.93.02090893.PMID2680588.S2CID7124561.Retrieved30 August2021.
  54. ^Ramírez-Venegas A, Torres-Duque CA, Guzmán-Bouilloud NE, González-García M, Sansores RH (2019)."SMALLa AIRWAY DISEASE IN COPD ASSOCIATED TO BIOMASS EXPOSURE".Rev Invest Clin.71(1): 70–78.doi:10.24875/RIC.18002652.PMID30810542.
  55. ^Corlateanu A, Mendez Y, Wang Y, Garnica RJ, Botnaru V, Siafakas N (2020)."Chronic obstructive pulmonary disease and phenotypes: a state-of-the-art".Pulmonology.26(2): 95–100.doi:10.1016/j.pulmoe.2019.10.006.PMID31740261.
  56. ^Postma DS, Rabe KF (24 September 2015). "The Asthma–COPD Overlap Syndrome".New England Journal of Medicine.373(13): 1241–1249.doi:10.1056/NEJMra1411863.PMID26398072.
  57. ^Mekov E, Nuñez A, Sin DD, Ichinose M, Rhee CK, Maselli DJ, Coté A, Suppli Ulrik C, Maltais F, Anzueto A, Miravitlles M (June 2021)."Update on Asthma–COPD Overlap (ACO): A Narrative Review".International Journal of Chronic Obstructive Pulmonary Disease.16:1783–1799.doi:10.2147/COPD.S312560.PMC8216660.PMID34168440.
  58. ^abHalpin DM, Miravitlles M, Metzdorf N, Celli B (2017)."Impact and prevention of severe exacerbations of COPD: a review of the evidence".Int J Chron Obstruct Pulmon Dis.12:2891–2908.doi:10.2147/COPD.S139470.PMC5638577.PMID29062228.
  59. ^abBrightling C, Greening N (August 2019)."Airway inflammation in COPD: progress to precision medicine"(PDF).Eur Respir J.54(2).doi:10.1183/13993003.00651-2019.PMID31073084.S2CID149444134.
  60. ^abcdeGold Report 2021,pp. 54–58, Chapter 3: Evidence supporting prevention and maintenance therapy.
  61. ^Bai S, Zhao L (2021)."Imbalance Between Injury and Defense in the COPD Emphysematous Phenotype".Frontiers in Medicine.8:653332.doi:10.3389/fmed.2021.653332.PMC8131650.PMID34026786.
  62. ^Kumar A, Mahajan A, Salazar EA, et al. (June 2021)."Impact of human immunodeficiency virus on pulmonary vascular disease".Glob Cardiol Sci Pract.2021(2): e202112.doi:10.21542/gcsp.2021.12.PMC8272407.PMID34285903.
  63. ^Osadnik CR, McDonald CF, Holland AE (2014)."Clinical issues of mucus accumulation in COPD".Int J Chron Obstruct Pulmon Dis.9:301–2.doi:10.2147/COPD.S61797.PMC3970915.PMID24741301.
  64. ^abSilverman EK (February 2020)."Genetics of COPD".Annu Rev Physiol.82:413–431.doi:10.1146/annurev-physiol-021317-121224.PMC7193187.PMID31730394.
  65. ^"FAQs Dust - Health and Safety Executive (HSE)".
  66. ^abNikolić MZ, Sun D, Rawlins EL (15 August 2018)."Human lung development: recent progress and new challenges".Development.145(16).doi:10.1242/dev.163485.PMC6124546.PMID30111617.
  67. ^Smith BM, Traboulsi H, Austin JM, et al. (January 2018)."Human airway branch variation and chronic obstructive pulmonary disease".Proc Natl Acad Sci U S A.115(5): E974–E981.Bibcode:2018PNAS..115E.974S.doi:10.1073/pnas.1715564115.PMC5798356.PMID29339516.
  68. ^abBailey KL, Samuelson DR, Wyatt TA (February 2021)."Alcohol use disorder: A pre-existing condition for COVID-19?".Alcohol.90:11–17.doi:10.1016/j.alcohol.2020.10.003.PMC7568767.PMID33080339.
  69. ^abSmith P, Jeffers LA, Koval M (November 2019)."Effects of different routes of endotoxin injury on barrier function in alcoholic lung syndrome".Alcohol.80:81–89.doi:10.1016/j.alcohol.2018.08.007.PMC6613986.PMID31278041.
  70. ^Arvers P (December 2018). "[Alcohol consumption and lung damage: Dangerous relationships]".Rev Mal Respir(in French).35(10): 1039–1049.doi:10.1016/j.rmr.2018.02.009.PMID29941207.S2CID239523761.
  71. ^Slovinsky WS, Romero F, Sales D, Shaghaghi H, Summer R (November 2019)."The involvement of GM-CSF deficiencies in parallel pathways of pulmonary alveolar proteinosis and the alcoholic lung".Alcohol.80:73–79.doi:10.1016/j.alcohol.2018.07.006.PMC6592783.PMID31229291.
  72. ^abWard H (2012).Oxford Handbook of Epidemiology for Clinicians.Oxford University Press. pp. 289–290.ISBN978-0-19-165478-7.
  73. ^Rennard S (2013).Clinical management of chronic obstructive pulmonary disease(2nd ed.). Informa Healthcare. p. 23.ISBN978-0-8493-7588-0.
  74. ^Sharma A, Barclay J (2010).COPD in primary care.Radcliffe Pub. p. 9.ISBN978-1-84619-316-3.
  75. ^Han MK, Martinez FJ (September 2020)."Host, Gender, and Early-Life Factors as Risks for Chronic Obstructive Pulmonary Disease".Clinics in Chest Medicine.41(3): 329–337.doi:10.1016/j.ccm.2020.06.009.PMC7993923.PMID32800188.
  76. ^Amaral AF, Strachan DP, Burney PG, Jarvis DL (May 2017)."Female Smokers Are at Greater Risk of Airflow Obstruction Than Male Smokers. UK Biobank"(PDF).American Journal of Respiratory and Critical Care Medicine.195(9): 1226–1235.doi:10.1164/rccm.201608-1545OC.hdl:10044/1/45106.PMID28075609.S2CID9360093.
  77. ^"Tobacco".Fact Sheets.World Health Organization.Retrieved12 July2021.
  78. ^Savran O, Ulrik CS (2018)."Early life insults as determinants of chronic obstructive pulmonary disease in adult life".International Journal of Chronic Obstructive Pulmonary Disease.13:683–693.doi:10.2147/COPD.S153555.PMC5834168.PMID29520136.
  79. ^Patel MP, Khangoora VS, Marik PE (October 2019). "A Review of the Pulmonary and Health Impacts of Hookah Use".Annals of the American Thoracic Society.16(10): 1215–1219.doi:10.1513/AnnalsATS.201902-129CME.PMID31091965.S2CID155103502.
  80. ^abcdChatkin JM, Zabert G, Zabert I, Chatkin G, Jiménez-Ruiz CA, de Granda-Orive JI, et al. (September 2017). "Lung Disease Associated With Marijuana Use".Arch Bronconeumol.53(9): 510–515.doi:10.1016/j.arbres.2017.03.019.PMID28483343.
  81. ^Underner M, Urban T, Perriot J, Peiffer G, Harika-Germaneau G, Jaafari N (December 2018). "[Spontaneous pneumothorax and lung emphysema in cannabis users]".Rev Pneumol Clin(in French).74(6): 400–415.doi:10.1016/j.pneumo.2018.06.003.PMID30420278.S2CID59233744.
  82. ^Martinasek MP, McGrogan JB, Maysonet A (November 2016)."A Systematic Review of the Respiratory Effects of Inhalational Marijuana".Respir Care.61(11): 1543–1551.doi:10.4187/respcare.04846.PMID27507173.
  83. ^Ribeiro LI, Ind PW (October 2016)."Effect of cannabis smoking on lung function and respiratory symptoms: a structured literature review".npj Primary Care Respiratory Medicine.26:16071.doi:10.1038/npjpcrm.2016.71.PMC5072387.PMID27763599.
  84. ^"Access to clean fuels and technologies for cooking".Our World in Data.Retrieved15 February2020.
  85. ^Halbert RJ, Natoli JL, Gano A, Badamgarav E, Buist AS, Mannino DM (September 2006)."Global burden of COPD: systematic review and meta-analysis".The European Respiratory Journal.28(3): 523–32.doi:10.1183/09031936.06.00124605.PMID16611654.
  86. ^abcdefghPirozzi C, Scholand MB (July 2012). "Smoking cessation and environmental hygiene".The Medical Clinics of North America.96(4): 849–67.doi:10.1016/j.mcna.2012.04.014.PMID22793948.
  87. ^Hopper T (2014).Mosby's Pharmacy Technician – E-Book: Principles and Practice.Elsevier Health Sciences. p. 610.ISBN978-0-323-29245-0.
  88. ^Barnes PJ, Drazen JM, Rennard SI (2009)."Relationship between cigarette smoking and occupational exposures".In Barnes PJ, Drazen JM, Rennard SI, Thomson NC (eds.).Asthma and COPD: Basic Mechanisms and Clinical Management.Academic. p. 464.ISBN978-0-12-374001-4.
  89. ^abForeman MG, Campos M, Celedón JC (July 2012)."Genes and chronic obstructive pulmonary disease".The Medical Clinics of North America.96(4): 699–711.doi:10.1016/j.mcna.2012.02.006.PMC3399759.PMID22793939.
  90. ^Brode SK, Ling SC, Chapman KR (September 2012)."Alpha-1 antitrypsin deficiency: a commonly overlooked cause of lung disease".CMAJ.184(12): 1365–71.doi:10.1503/cmaj.111749.PMC3447047.PMID22761482.
  91. ^abcdeReilly JJ, Silverman EK, Shapiro SD (2011). "Chronic Obstructive Pulmonary Disease". In Longo D, Fauci A, Kasper D, Hauser S, Jameson J, Loscalzo J (eds.).Harrison's Principles of Internal Medicine(18th ed.). McGraw Hill. pp. 2151–9.ISBN978-0-07-174889-6.
  92. ^"Home - Gene - NCBI".ncbi.nlm.nih.gov.Retrieved17 July2022.
  93. ^Maselli DJ, Bhatt SP, Anzueto A, Bowler RP, DeMeo DL, Diaz AA, et al. (August 2019)."Clinical Epidemiology of COPD: Insights From 10 Years of the COPDGene Study".Chest.156(2): 228–238.doi:10.1016/j.chest.2019.04.135.PMC7198872.PMID31154041.
  94. ^Capron T, Bourdin A, Perez T, Chanez P (June 2019)."COPD beyond proximal bronchial obstruction: phenotyping and related tools at the bedside"(PDF).Eur Respir Rev.28(152): 190010.doi:10.1183/16000617.0010-2019.PMC9488991.PMID31285287.S2CID195844108.
  95. ^Kumar V, Abbas AK, Aster JC (2018).Robbins basic pathology(10th ed.). Elsevier. pp. 498–502.ISBN978-0-323-35317-5.
  96. ^abD'Ascanio M, Viccaro F, Calabrò N, Guerrieri G, Salvucci C, Pizzirusso D, et al. (2020)."Assessing Static Lung Hyperinflation by Whole-Body Plethysmography, Helium Dilution, and Impulse Oscillometry System (IOS) in Patients with COPD".Int J Chron Obstruct Pulmon Dis.15:2583–9.doi:10.2147/COPD.S264261.PMC7585810.PMID33116475.
  97. ^Knox-Brown B, Patel J, Potts J, Ahmed R, Aquart-Stewart A, Cherkaski H, et al. (January 2023). "Small airways obstruction and its risk factors in the Burden of Obstructive Lung Disease (BOLD) study: a multinational cross-sectional study".Lancet Glob Health.11(1): e69–e82.doi:10.1016/S2214-109X(22)00456-9.hdl:10044/1/100800.PMID36521955.S2CID254665705.
  98. ^Knox-Brown B, Patel J, Potts J, et al. (23 May 2023)."The association of spirometric small airways obstruction with respiratory symptoms, cardiometabolic diseases, and quality of life: results from the Burden of Obstructive Lung Disease (BOLD) study".Respiratory Research.24(1): 137.doi:10.1186/s12931-023-02450-1.PMC10207810.PMID37221593.
  99. ^Knox-Brown B, Potts J, Santofimio VQ, Minelli C, Patel J, Abass NM, Agarwal D, Ahmed R, Mahesh PA, Bs J, Denguezli M, Franssen F, Gislason T, Janson C, Juvekar SK, Koul P, Malinovschi A, Nafees AA, Nielsen R, Paraguas SN, Buist S, Burney PG, Amaral AF (November 2023). "Isolated small airways obstruction predicts future chronic airflow obstruction: a multinational longitudinal study".BMJ Open Respiratory Research.10(1): e002056.doi:10.1136/bmjresp-2023-002056.hdl:20.500.11815/4593.
  100. ^Lo Bello F, Ieni A, Hansbro PM, Ruggeri P, Di Stefano A, Nucera F, et al. (May 2020). "Role of the mucins in pathogenesis of COPD: implications for therapy".Expert Review of Respiratory Medicine.14(5): 465–483.doi:10.1080/17476348.2020.1739525.hdl:10453/139160.PMID32133884.S2CID212416323.
  101. ^abHigham A, Quinn AM, Cançado JE, Singh D (March 2019)."The pathology of small airways disease in COPD: historical aspects and future directions".Respir Res.20(1): 49.doi:10.1186/s12931-019-1017-y.PMC6399904.PMID30832670.
  102. ^abCalverley PM, Koulouris NG (January 2005)."Flow limitation and dynamic hyperinflation: key concepts in modern respiratory physiology".The European Respiratory Journal.25(1): 186–99.doi:10.1183/09031936.04.00113204.PMID15640341.
  103. ^Currie GP (2010).ABC of COPD(2nd ed.). Wiley-Blackwell, BMJ Books. p. 32.ISBN978-1-4443-2948-3.
  104. ^O'Donnell DE (April 2006). "Hyperinflation, dyspnea, and exercise intolerance in chronic obstructive pulmonary disease".Proceedings of the American Thoracic Society.3(2): 180–4.doi:10.1513/pats.200508-093DO.PMID16565429.S2CID20644418.
  105. ^Cooper CB (October 2006). "The connection between chronic obstructive pulmonary disease symptoms and hyperinflation and its impact on exercise and function".The American Journal of Medicine.119(10 Suppl 1): 21–31.doi:10.1016/j.amjmed.2006.08.004.PMID16996896.
  106. ^abc"Chronic obstructive pulmonary disease - NICE Pathways".pathways.nice.org.uk.9 April 2020.Retrieved29 June2021.
  107. ^abcdQaseem A, Wilt TJ, Weinberger SE, Hanania NA, Criner G, van der Molen T, et al. (August 2011). "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".Annals of Internal Medicine.155(3): 179–191.doi:10.7326/0003-4819-155-3-201108020-00008.PMID21810710.S2CID18830625.
  108. ^abYoung VB (2010).Blueprints medicine(5th ed.). Wolters Kluwer Health/Lippincott Williams & Wilkins. p. 69.ISBN978-0-7817-8870-0.
  109. ^abcNational Institute for Health and Clinical Excellence.Clinical guideline 101: Chronic Obstructive Pulmonary Disease.London, June 2010.
  110. ^Bailey KL (July 2012)."The importance of the assessment of pulmonary function in COPD".The Medical Clinics of North America.96(4): 745–752.doi:10.1016/j.mcna.2012.04.011.PMC3998207.PMID22793942.
  111. ^Williams N (August 2017)."The MRC breathlessness scale".Occup Med (Lond).67(6): 496–497.doi:10.1093/occmed/kqx086.PMID28898975.
  112. ^ab"Chronic obstructive pulmonary disease (COPD) - Criteria | BMJ Best Practice".bestpractice.bmj.Retrieved2 July2021.
  113. ^"COPD Assessment Test (CAT)".American Thoracic Society.Archivedfrom the original on December 3, 2013.RetrievedNovember 29,2013.
  114. ^Tyagi J, Moola S, Bhaumik S (June 2021)."Diagnostic accuracy of screening tools for chronic obstructive pulmonary disease in primary health care: Rapid evidence synthesis".Journal of Family Medicine and Primary Care.10(6): 2184–2194.doi:10.4103/jfmpc.jfmpc_2263_20.PMC8284240.PMID34322411.
  115. ^"saber sheath trachea | Search | Radiopaedia.org".Radiopaedia.Retrieved13 August2021.
  116. ^abVestbo J, Hurd SS, Agustí AG, et al. (February 2013)."Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary".American Journal of Respiratory and Critical Care Medicine.187(4): 350–352.doi:10.1164/rccm.201204-0596PP.PMID22878278.
  117. ^BTS COPD Consortium (2005)."Spirometry in practice – a practical guide to using spirometry in primary care".pp. 8–9. Archived fromthe originalon 26 August 2014.Retrieved25 August2014.
  118. ^abcPolicy Recommendations for Smoking Cessation and Treatment of Tobacco Dependence.World Health Organization. 2003. pp. 15–40.ISBN978-92-4-156240-9.Archived fromthe originalon 2008-09-15.
  119. ^Jiménez-Ruiz CA, Fagerström KO (March 2013)."Smoking cessation treatment for COPD smokers: the role of counselling".Monaldi Archives for Chest Disease.79(1): 33–7.doi:10.4081/monaldi.2013.107.PMID23741944.
  120. ^ab"Chronic obstructive pulmonary disease in over 16s: diagnosis and management | Guidance and guidelines | NICE".nice.org.uk.23 June 2010.Retrieved2018-06-05.
  121. ^Kumar P, Clark M (2005).Clinical Medicine(6th ed.). Elsevier Saunders. pp. 900–1.ISBN978-0-7020-2763-5.
  122. ^abTønnesen P (March 2013)."Smoking cessation and COPD".European Respiratory Review.22(127): 37–43.doi:10.1183/09059180.00007212.PMC9487432.PMID23457163.
  123. ^"Coping with cravings".nhs.uk.27 April 2018.Retrieved15 July2021.
  124. ^van Eerd EA, van der Meer RM, van Schayck OC, Kotz D (August 2016)."Smoking cessation for people with chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2019(8): CD010744.doi:10.1002/14651858.CD010744.pub2.PMC6400424.PMID27545342.
  125. ^Smith BK, Timby NE (2005).Essentials of nursing: care of adults and children.Lippincott Williams & Wilkins. p. 338.ISBN978-0-7817-5098-1.
  126. ^Rom WN, Markowitz SB, eds. (2007).Environmental and occupational medicine(4th ed.). Wolters Kluwer/Lippincott Williams & Wilkins. pp. 521–522.ISBN978-0-7817-6299-1.
  127. ^"Wet cutting".Health and Safety Executive.Archivedfrom the original on December 3, 2013.RetrievedNovember 29,2013.
  128. ^George RB (2005).Chest medicine: essentials of pulmonary and critical care medicine(5th ed.). Lippincott Williams & Wilkins. p. 172.ISBN978-0-7817-5273-2.
  129. ^abcdefghijVestbo J, Hurd SS, Agustí AG, Jones PW, Vogelmeier C, Anzueto A, et al. (February 2013)."Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease: GOLD executive summary".American Journal of Respiratory and Critical Care Medicine.187(4): 347–365.doi:10.1164/rccm.201204-0596PP.PMID22878278.
  130. ^"Chronic obstructive pulmonary disease (COPD) - Treatment - NHS".20 October 2017.
  131. ^CDC (2024-01-18)."RSV information for healthcare providers".Centers for Disease Control and Prevention.Retrieved2024-02-23.
  132. ^"2024 GOLD Report".Global Initiative for Chronic Obstructive Lung Disease - GOLD.Retrieved2024-02-23.
  133. ^Howcroft M, Walters EH, Wood-Baker R, Walters JA (December 2016)."Action plans with brief patient education for exacerbations in chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2016(12): CD005074.doi:10.1002/14651858.CD005074.pub4.PMC6463844.PMID27990628.
  134. ^Lenferink A, Brusse-Keizer M, van der Valk PD, Frith PA, Zwerink M, Monninkhof EM, et al. (August 2017)."Self-management interventions including action plans for exacerbations versus usual care in patients with chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2017(8): CD011682.doi:10.1002/14651858.CD011682.pub2.PMC6483374.PMID28777450.
  135. ^Jolliffe DA, Greenberg L, Hooper RL, Mathyssen C, Rafiq R, de Jongh RT, et al. (April 2019)."Vitamin D to prevent exacerbations of COPD: systematic review and meta-analysis of individual participant data from randomised controlled trials".Thorax.74(4): 337–345.doi:10.1136/thoraxjnl-2018-212092.PMID30630893.
  136. ^abVollenweider DJ, Frei A, Steurer-Stey CA, Garcia-Aymerich J, Puhan MA (October 2018)."Antibiotics for exacerbations of chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2018(10): CD010257.doi:10.1002/14651858.CD010257.pub2.PMC6517133.PMID30371937.
  137. ^Mackay AJ, Hurst JR (July 2012). "COPD exacerbations: causes, prevention, and treatment".The Medical Clinics of North America.96(4): 789–809.doi:10.1016/j.mcna.2012.02.008.PMID22793945.
  138. ^"Fluoroquinolone Antibacterial Drugs: Drug Safety Communication – FDA Advises Restricting Use for Certain Uncomplicated Infections".FDA.12 May 2016.Archivedfrom the original on 16 May 2016.Retrieved16 May2016.
  139. ^abcdefghDecramer M, Janssens W, Miravitlles M (April 2012)."Chronic obstructive pulmonary disease".Lancet.379(9823): 1341–51.CiteSeerX10.1.1.1000.1967.doi:10.1016/S0140-6736(11)60968-9.PMC7172377.PMID22314182.
  140. ^Oliveira EP, Medeiros Junior P (2020)."Palliative care in pulmonary medicine".Jornal Brasileiro de Pneumologia(in Portuguese).46(3): e20190280.doi:10.36416/1806-3756/e20190280.PMC7572288.PMID32638839.
  141. ^Wilson ME, Dobler CC, Morrow AS, Beuschel B, Alsawas M, Benkhadra R, et al. (February 2020)."Association of Home Noninvasive Positive Pressure Ventilation With Clinical Outcomes in Chronic Obstructive Pulmonary Disease: A Systematic Review and Meta-analysis".JAMA.323(5): 455–465.doi:10.1001/jama.2019.22343.PMC7042860.PMID32016309.
  142. ^Almadhoun K, Sharma S (2021)."Bronchodilators".StatPearls.StatPearls Publishing.PMID30085570.Retrieved27 July2021.
  143. ^abcGold Report 2021,pp. 48–52, Chapter 3: Evidence supporting prevention and maintenance therapy.
  144. ^Farne HA, Cates CJ (October 2015)."Long-acting beta2-agonist in addition to tiotropium versus either tiotropium or long-acting beta2-agonist alone for chronic obstructive pulmonary disease"(PDF).The Cochrane Database of Systematic Reviews.2015(10): CD008989.doi:10.1002/14651858.CD008989.pub3.PMC4164463.PMID26490945.
  145. ^Oba Y, Keeney E, Ghatehorde N, Dias S, et al. (Cochrane Airways Group) (December 2018)."Dual combination therapy versus long-acting bronchodilators alone for chronic obstructive pulmonary disease (COPD): a systematic review and network meta-analysis".The Cochrane Database of Systematic Reviews.2018(12): CD012620.doi:10.1002/14651858.CD012620.pub2.PMC6517098.PMID30521694.
  146. ^ab"Recommendations | Chronic obstructive pulmonary disease in over 16s: diagnosis and management | Guidance | NICE".nice.org.uk.5 December 2018.Retrieved1 August2021.
  147. ^abCave AC, Hurst MM (May 2011). "The use of long acting β₂-agonists, alone or in combination with inhaled corticosteroids, in chronic obstructive pulmonary disease (COPD): a risk-benefit analysis".Pharmacology & Therapeutics.130(2): 114–143.doi:10.1016/j.pharmthera.2010.12.008.PMID21276815.
  148. ^Spencer S, Karner C, Cates CJ, Evans DJ (December 2011). Spencer S (ed.)."Inhaled corticosteroids versus long-acting beta(2)-agonists for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2011(12): CD007033.doi:10.1002/14651858.CD007033.pub3.PMC6494276.PMID22161409.
  149. ^Wang J, Nie B, Xiong W, Xu Y (April 2012)."Effect of long-acting beta-agonists on the frequency of COPD exacerbations: a meta-analysis".Journal of Clinical Pharmacy and Therapeutics.37(2): 204–211.doi:10.1111/j.1365-2710.2011.01285.x.PMID21740451.S2CID45383688.
  150. ^abGeake JB, Dabscheck EJ, Wood-Baker R, Cates CJ (January 2015)."Indacaterol, a once-daily beta2-agonist, versus twice-daily beta₂-agonists or placebo for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.1(3): CD010139.doi:10.1002/14651858.CD010139.pub2.PMC6464646.PMID25575340.
  151. ^Decramer ML, Hanania NA, Lötvall JO, Yawn BP (2013)."The safety of long-acting β2-agonists in the treatment of stable chronic obstructive pulmonary disease".International Journal of Chronic Obstructive Pulmonary Disease.8:53–64.doi:10.2147/COPD.S39018.PMC3558319.PMID23378756.
  152. ^Nannini LJ, Lasserson TJ, Poole P (September 2012). Nannini LJ (ed.)."Combined corticosteroid and long-acting beta(2)-agonist in one inhaler versus long-acting beta(2)-agonists for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.9(9): CD006829.doi:10.1002/14651858.CD006829.pub2.PMC4170910.PMID22972099.
  153. ^Fukuda N, Horita N, Kaneko A, Goto A, Kaneko T, Ota E, Kew KM (2023-06-05)."Long-acting muscarinic antagonist (LAMA) plus long-acting beta-agonist (LABA) versus LABA plus inhaled corticosteroid (ICS) for stable chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2023(6): CD012066.doi:10.1002/14651858.CD012066.pub3.ISSN1469-493X.PMC10241721.PMID37276335.
  154. ^Zheng Y, Zhu J, Liu Y, Lai W, Lin C, Qiu K, et al. (November 2018)."Triple therapy in the management of chronic obstructive pulmonary disease: systematic review and meta-analysis".BMJ.363:k4388.doi:10.1136/bmj.k4388.PMC6218838.PMID30401700.
  155. ^abKarner C, Chong J, Poole P (July 2014)."Tiotropium versus placebo for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2016(7): CD009285.doi:10.1002/14651858.CD009285.pub3.PMC8934583.PMID25046211.
  156. ^Cheyne L, Irvin-Sellers MJ, White J (September 2015)."Tiotropium versus ipratropium bromide for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2015(9): CD009552.doi:10.1002/14651858.CD009552.pub3.PMC8749963.PMID26391969.
  157. ^Singh S, Loke YK, Enright P, Furberg CD (January 2013)."Pro-arrhythmic and pro-ischaemic effects of inhaled anticholinergic medications".Thorax.68(1): 114–6.doi:10.1136/thoraxjnl-2011-201275.PMID22764216.
  158. ^Jones P (April 2013). "Aclidinium bromide twice daily for the treatment of chronic obstructive pulmonary disease: a review".Advances in Therapy.30(4): 354–68.doi:10.1007/s12325-013-0019-2.PMID23553509.S2CID3530290.
  159. ^Cazzola M, Page CP, Matera MG (June 2013). "Aclidinium bromide for the treatment of chronic obstructive pulmonary disease".Expert Opinion on Pharmacotherapy.14(9): 1205–14.doi:10.1517/14656566.2013.789021.PMID23566013.S2CID24973904.
  160. ^abNi H, Soe Z, Moe S (September 2014)."Aclidinium bromide for stable chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.9(9): CD010509.doi:10.1002/14651858.CD010509.pub2.PMC8922974.PMID25234126.
  161. ^Ni H, Htet A, Moe S (June 2017)."Umeclidinium bromide versus placebo for people with chronic obstructive pulmonary disease (COPD)".The Cochrane Database of Systematic Reviews.2017(6): CD011897.doi:10.1002/14651858.CD011897.pub2.PMC6481854.PMID28631387.
  162. ^Ismaila AS, Huisman EL, Punekar YS, Karabis A (2015)."Comparative efficacy of long-acting muscarinic antagonist monotherapies in COPD: a systematic review and network meta-analysis".International Journal of Chronic Obstructive Pulmonary Disease.10:2495–517.doi:10.2147/COPD.S92412.PMC4655912.PMID26604738.
  163. ^abChen H, Sun J, Huang Q, Liu Y, Yuan M, Ma C, et al. (2021)."Inhaled Corticosteroids and the Pneumonia Risk in Patients With Chronic Obstructive Pulmonary Disease: A Meta-analysis of Randomized Controlled Trials".Front Pharmacol.12:691621.doi:10.3389/fphar.2021.691621.PMC8275837.PMID34267661.
  164. ^Chinet T, Dumoulin J, Honore I, Braun JM, Couderc LJ, Febvre M, et al. (December 2016). "[The place of inhaled corticosteroids in COPD]".Revue des Maladies Respiratoires.33(10): 877–891.doi:10.1016/j.rmr.2015.11.009.PMID26831345.
  165. ^Walters JA, Tan DJ, White CJ, Gibson PG, Wood-Baker R, Walters EH (September 2014)."Systemic corticosteroids for acute exacerbations of chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.9(9): CD001288.doi:10.1002/14651858.CD001288.pub4.PMC11195634.PMID25178099.
  166. ^Walters JA, Tan DJ, White CJ, Wood-Baker R (March 2018)."Different durations of corticosteroid therapy for exacerbations of chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2018(3): CD006897.doi:10.1002/14651858.CD006897.pub4.PMC6494402.PMID29553157.
  167. ^van Geffen WH, Tan DJ, Walters JA, Walters EH (2023-12-06). Cochrane Airways Group (ed.). "Inhaled corticosteroids with combination inhaled long-acting beta2-agonists and long-acting muscarinic antagonists for chronic obstructive pulmonary disease".Cochrane Database of Systematic Reviews.2023(12): CD011600.doi:10.1002/14651858.CD011600.pub3.PMC10698842.PMID38054551.
  168. ^Mammen MJ, Sethi S (2012). "Macrolide therapy for the prevention of acute exacerbations in chronic obstructive pulmonary disease".Polskie Archiwum Medycyny Wewnetrznej.122(1–2): 54–9.doi:10.20452/pamw.1134.PMID22353707.S2CID35183033.
  169. ^abHerath SC, Normansell R, Maisey S, Poole P (October 2018)."Prophylactic antibiotic therapy for chronic obstructive pulmonary disease (COPD)".The Cochrane Database of Systematic Reviews.2018(10): CD009764.doi:10.1002/14651858.CD009764.pub3.PMC6517028.PMID30376188.
  170. ^Simoens S, Laekeman G, Decramer M (May 2013)."Preventing COPD exacerbations with macrolides: a review and budget impact analysis".Respiratory Medicine.107(5): 637–48.doi:10.1016/j.rmed.2012.12.019.PMID23352223.
  171. ^Poole P, Sathananthan K, Fortescue R (May 2019)."Mucolytic agents versus placebo for chronic bronchitis or chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.5(3): CD001287.doi:10.1002/14651858.CD001287.pub6.PMC6527426.PMID31107966.
  172. ^"Erdosteine".NICE.Retrieved20 July2021.
  173. ^Meldrum OW, Chotirmall SH (June 2021)."Mucus, Microbiomes and Pulmonary Disease".Biomedicines.9(6): 675.doi:10.3390/biomedicines9060675.PMC8232003.PMID34199312.
  174. ^"Bronchitis".nhs.uk.17 October 2017.
  175. ^Zhu A, Teng Y, Ge D, Zhang X, Hu M, Yao X (October 2019)."Role of metformin in treatment of patients with chronic obstructive pulmonary disease: a systematic review".Journal of Thoracic Disease.11(10): 4371–4378.doi:10.21037/jtd.2019.09.84.PMC6837976.PMID31737323.
  176. ^abKhor YH, Renzoni EA, Visca D, McDonald CF, Goh NS (July 2019)."Oxygen therapy in COPD and interstitial lung disease: navigating the knowns and unknowns".ERJ Open Res.5(3).doi:10.1183/23120541.00118-2019.PMC6745413.PMID31544111.
  177. ^Ekström M, Ahmadi Z, Bornefalk-Hermansson A, Abernethy A, Currow D (November 2016)."Oxygen for breathlessness in patients with chronic obstructive pulmonary disease who do not qualify for home oxygen therapy".The Cochrane Database of Systematic Reviews.11(8): CD006429.doi:10.1002/14651858.CD006429.pub3.PMC6464154.PMID27886372.
  178. ^Jindal SK (2013).Chronic Obstructive Pulmonary Disease.Jaypee Brothers Medical. p. 139.ISBN978-93-5090-353-7.
  179. ^abO'Driscoll BR, Howard LS, Davison AG (October 2008)."BTS guideline for emergency oxygen use in adult patients".Thorax.63(6): vi1-68.doi:10.1136/thx.2008.102947.PMID18838559.
  180. ^"2024 GOLD Report".Global Initiative for Chronic Obstructive Lung Disease - GOLD.Retrieved2024-02-23.
  181. ^Venkatesan P (January 2024)."GOLD COPD report: 2024 update".The Lancet Respiratory Medicine.12(1): 15–16.doi:10.1016/S2213-2600(23)00461-7.PMID38061380.S2CID265751322.
  182. ^"Pulmonary Rehabilitation".medlineplus.gov.Retrieved9 September2021.
  183. ^McNamara RJ, McKeough ZJ, McKenzie DK, Alison JA (December 2013). "Water-based exercise training for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews(12): CD008290.doi:10.1002/14651858.CD008290.pub2.PMID24353107.
  184. ^Menadue C, Piper AJ, van 't Hul AJ, Wong KK (May 2014)."Non-invasive ventilation during exercise training for people with chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.2014(5): CD007714.doi:10.1002/14651858.CD007714.pub2.PMC10984247.PMID24823712.
  185. ^abMcKeough ZJ, Velloso M, Lima VP, Alison JA (November 2016)."Upper limb exercise training for COPD".The Cochrane Database of Systematic Reviews.2016(11): CD011434.doi:10.1002/14651858.CD011434.pub2.PMC6464968.PMID27846347.
  186. ^abNgai SP, Jones AY, Tam WW (June 2016)."Tai Chi for chronic obstructive pulmonary disease (COPD)".The Cochrane Database of Systematic Reviews.2016(6): CD009953.doi:10.1002/14651858.CD009953.pub2.PMC8504989.PMID27272131.
  187. ^Ammous O, Feki W, Lotfi T, Khamis AM, Gosselink R, Rebai A, Kammoun S (2023-01-06). Cochrane Airways Group (ed.)."Inspiratory muscle training, with or without concomitant pulmonary rehabilitation, for chronic obstructive pulmonary disease (COPD)".Cochrane Database of Systematic Reviews.2023(1): CD013778.doi:10.1002/14651858.CD013778.pub2.PMC9817429.PMID36606682.
  188. ^Thomas MJ, Simpson J, Riley R, Grant E (June 2010). "The impact of home-based physiotherapy interventions on breathlessness during activities of daily living in severe COPD: a systematic review".Physiotherapy.96(2): 108–19.doi:10.1016/j.physio.2009.09.006.PMID20420957.
  189. ^abWearing J, Beaumont S, Forbes D, Brown B, Engel R (February 2016)."The Use of Spinal Manipulative Therapy in the Management of Chronic Obstructive Pulmonary Disease: A Systematic Review".Journal of Alternative and Complementary Medicine.22(2): 108–14.doi:10.1089/acm.2015.0199.PMC4761829.PMID26700633.
  190. ^Simonelli C, Vitacca M, Vignoni M, Ambrosino N, Paneroni M (2019)."Effectiveness of manual therapy in COPD: A systematic review of randomised controlled trials".Pulmonology.25(4): 236–247.doi:10.1016/j.pulmoe.2018.12.008.PMID30738792.
  191. ^abOsadnik CR, McDonald CF, Jones AP, Holland AE (March 2012)."Airway clearance techniques for chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.3(3): CD008328.doi:10.1002/14651858.CD008328.pub2.PMC11285303.PMID22419331.
  192. ^Ferreira IM, Brooks D, White J, Goldstein R (December 2012). Ferreira IM (ed.). "Nutritional supplementation for stable chronic obstructive pulmonary disease".The Cochrane Database of Systematic Reviews.12:CD000998.doi:10.1002/14651858.CD000998.pub3.PMID23235577.
  193. ^Van Geffen WH, Douma WR, Slebos DJ, Kerstjens HA (29 August 2016)."Bronchodilators delivered by nebuliser versus inhalers for lung attacks of chronic obstructive pulmonary disease".Cochrane Database of Systematic Reviews.29(8): 011826.doi:10.1002/14651858.CD011826.pub2.hdl:11370/95fc3e6e-ebd0-440f-9721-489729f80add.PMC8487315.PMID27569680.
  194. ^Wise R."Chronic Obstructive Pulmonary Disease (COPD)".Pulmonary Disorders: Merck Manuals Professional Edition.Archivedfrom the original on 28 December 2016.Retrieved16 December2016.
  195. ^Osman LM, Ayres JG, Garden C, Reglitz K, Lyon J, Douglas JG (2008)."Home warmth and health status of COPD patients".Eur. J. Public Health.18(4): 399–405.doi:10.1093/eurpub/ckn015.PMID18367496.
  196. ^Mu Z, Chen PL, Geng FH, Ren L, Gu WC, Ma JY, Peng L, Li QY (2017). "Synergistic effects of temperature and humidity on the symptoms of COPD patients".Int J Biometeorol.61(11): 1919–25.Bibcode:2017IJBm...61.1919M.doi:10.1007/s00484-017-1379-0.PMID28567499.S2CID25962322.
  197. ^"Humidifiers: Ease skin, breathing symptoms".Mayo Clinic.
  198. ^Noti JD, Blachere FM, McMillen CM, Lindsley WG, Kashon ML, Slaughter DR, Beezhold DH (2013)."High humidity leads to loss of infectious influenza virus from simulated coughs".PLOS ONE.8(2): e57485.Bibcode:2013PLoSO...857485N.doi:10.1371/journal.pone.0057485.PMC3583861.PMID23460865.
  199. ^abGold Report 2021,pp. 92–96, Chapter 4: Management of stable COPD.
  200. ^van Geffen WH, Slebos DJ, Herth FJ, Kemp SV, Weder W, Shah PL (April 2019)."Surgical and endoscopic interventions that reduce lung volume for emphysema: a systemic review and meta-analysis"(PDF).The Lancet. Respiratory Medicine.7(4): 313–324.doi:10.1016/S2213-2600(18)30431-4.PMID30744937.S2CID73428098.
  201. ^abDuffy S, Marchetti N, Criner GJ (September 2020). "Surgical Therapies for Chronic Obstructive Pulmonary Disease".Clinics in Chest Medicine.41(3): 559–566.doi:10.1016/j.ccm.2020.06.011.PMID32800206.S2CID221145423.
  202. ^ab"1 Recommendations | Endobronchial valve insertion to reduce lung volume in emphysema | Guidance | NICE".nice.org.uk.20 December 2017.Retrieved7 July2021.
  203. ^Klooster K, Slebos DJ (May 2021)."Endobronchial Valves for the Treatment of Advanced Emphysema".Chest.159(5): 1833–1842.doi:10.1016/j.chest.2020.12.007.PMC8129734.PMID33345947.
  204. ^Welling JB, Slebos DJ (August 2018)."Lung volume reduction with endobronchial coils for patients with emphysema".J Thorac Dis.10(Suppl 23): S2797–S2805.doi:10.21037/jtd.2017.12.95.PMC6129816.PMID30210833.
  205. ^Valipour A (January 2017). "Bronchoscopic Thermal Vapour Ablation: Hot Stuff to Treat Emphysema Patients!".Archivos de Bronconeumologia.53(1): 1–2.doi:10.1016/j.arbr.2016.11.009.PMID27916315.S2CID78181696.
  206. ^Mortensen J, Berg RM (January 2019)."Lung Scintigraphy in COPD".Seminars in Nuclear Medicine.49(1): 16–21.doi:10.1053/j.semnuclmed.2018.10.010.PMID30545511.S2CID56486118.
  207. ^"WHO Disease and injury country estimates".World Health Organization.2009.Archivedfrom the original on 2009-11-11.RetrievedNov 11,2009.
  208. ^abMurray CJ, Vos T, Lozano R, Naghavi M, Flaxman AD, Michaud C, et al. (December 2012). "Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010".Lancet.380(9859): 2197–2223.doi:10.1016/S0140-6736(12)61689-4.PMID23245608.S2CID205967479.
  209. ^Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. (December 2012)."Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010".Lancet.380(9859): 2163–2196.doi:10.1016/S0140-6736(12)61729-2.PMC6350784.PMID23245607.
  210. ^Gold Report 2021,pp. 26–33, Chapter 2:Diagnosis and assessment.
  211. ^ab"COPD prevalence".NICE.Retrieved18 July2021.
  212. ^Rycroft CE, Heyes A, Lanza L, Becker K (2012)."Epidemiology of chronic obstructive pulmonary disease: a literature review".International Journal of Chronic Obstructive Pulmonary Disease.7:457–94.doi:10.2147/COPD.S32330.PMC3422122.PMID22927753.
  213. ^Martinez FJ, Curtis JL, Sciurba F, Mumford J, Giardino ND, Weinmann G, Kazerooni E, Murray S, Criner GJ, Sin DD, Hogg J, Ries AL, Han M, Fishman AP, Make B, Hoffman EA, Mohsenifar Z, Wise R (April 2007)."Sex differences in severe pulmonary emphysema".American Journal of Respiratory and Critical Care Medicine.176(3): 243–252.doi:10.1164/rccm.200606-828OC.PMC1994221.PMID17431226.
  214. ^"Basics About COPD".Chronic Obstructive Pulmonary Disease (COPD).Center for Disease Control. 9 June 2021.Retrieved18 July2021.
  215. ^abTorio CM, Andrews RM (2006)."National Inpatient Hospital Costs: The Most Expensive Conditions by Payer, 2011: Statistical Brief #160".Healthcare Cost and Utilization Project (HCUP) Statistical Briefs.PMID24199255.Archivedfrom the original on 2017-03-14.
  216. ^"The top 10 causes of death".Fact Sheets.World Health Organization.
  217. ^abcdefPetty TL (2006)."The history of COPD".International Journal of Chronic Obstructive Pulmonary Disease.1(1): 3–14.doi:10.2147/copd.2006.1.1.3.PMC2706597.PMID18046898.
  218. ^abWright JL, Churg A (2008)."Pathologic Features of Chronic Obstructive Pulmonary Disease: Diagnostic Criteria and Differential Diagnosis"(PDF).In Fishman A, Elias J, Fishman J, Grippi M, Senior R, Pack A (eds.).Fishman's Pulmonary Diseases and Disorders(4th ed.). McGraw-Hill. pp. 693–705.ISBN978-0-07-164109-8.Archived fromthe original(PDF)on 2016-03-03.Retrieved2008-11-11.
  219. ^Woolcock A (1984). "The Search for Words to Describe the Bad Blowers".Chest.85(6): 73S–74S.doi:10.1378/chest.85.6_Supplement.73S.
  220. ^Waldbott GL (1965).A struggle with Titans.Carlton Press. p. 6.
  221. ^Fishman AP (May 2005). "One hundred years of chronic obstructive pulmonary disease".American Journal of Respiratory and Critical Care Medicine.171(9): 941–8.doi:10.1164/rccm.200412-1685OE.PMID15849329.
  222. ^"November is National COPD Awareness Month | NHLBI, NIH".nhlbi.nih.gov.Retrieved21 July2021.
  223. ^abLomborg B (2013).Global problems, local solutions: costs and benefits.Cambridge University Press. p. 143.ISBN978-1-107-03959-9.
  224. ^Bloom D (2011).The Global Economic Burden of Noncommunicable Diseases(PDF).World Economic Forum. p. 24.Archived(PDF)from the original on 2015-02-04.
  225. ^"NIH study shows hyaluronan is effective in treating chronic lung disease".National Institutes of Health (NIH).1 February 2021.Retrieved7 August2021.
  226. ^Poggi C, Mantovani S, Pecoraro Y, Carillo C, Bassi M, D'Andrilli A, Anile M, Rendina EA, Venuta F, Diso D (November 2018)."Bronchoscopic treatment of emphysema: an update".J Thorac Dis.10(11): 6274–6284.doi:10.21037/jtd.2018.10.43.PMC6297441.PMID30622803.
  227. ^"A Prospective Safety and Feasibility Study of the RejuvenAir™ System Metered Cryospray Therapy for Chronic Bronchitis Patients".clinicaltrials.gov. 25 January 2021.Retrieved16 August2021.
  228. ^Chen YT, Miao K, Zhou L, Xiong WN (June 2021)."Stem cell therapy for chronic obstructive pulmonary disease".Chin Med J (Engl).134(13): 1535–1545.doi:10.1097/CM9.0000000000001596.PMC8280064.PMID34250959.
  229. ^"First UK patients get pioneering new treatment for serious lung disease | Royal Brompton & Harefield hospitals".rbht.nhs.uk.Retrieved4 August2021.
  230. ^Gøtzsche PC, Johansen HK (September 2016)."Intravenous Alpha -1 antitrypsin augmentation therapy for treating patients with Alpha -1 antitrypsin deficiency and lung disease".The Cochrane Database of Systematic Reviews.2016(9): CD007851.doi:10.1002/14651858.CD007851.pub3.PMC6457738.PMID27644166.
  231. ^Campos MA, Geraghty P, Holt G, Mendes E, Newby PR, Ma S, et al. (August 2019)."The Biological Effects of Double-Dose Alpha-1 Antitrypsin Augmentation Therapy. A Pilot Clinical Trial".American Journal of Respiratory and Critical Care Medicine.200(3): 318–326.doi:10.1164/rccm.201901-0010OC.PMC6680306.PMID30965011.
  232. ^Nambiar S, Bong How S, Gummer J, Trengove R, Moodley Y (February 2020)."Metabolomics in chronic lung diseases".Respirology.25(2): 139–148.doi:10.1111/resp.13530.PMID30907495.
  233. ^McLean S, Nurmatov U, Liu JL, Pagliari C, Car J, Sheikh A (July 2011)."Telehealthcare for chronic obstructive pulmonary disease"(PDF).The Cochrane Database of Systematic Reviews.2012(7): CD007718.doi:10.1002/14651858.CD007718.pub2.PMC8939044.PMID21735417.
  234. ^Cheng J, Eroglu A (June 2021)."The Promising Effects of Astaxanthin on Lung Diseases".Adv Nutr.12(3): 850–864.doi:10.1093/advances/nmaa143.PMC8166543.PMID33179051.
  235. ^Gruß I, McCreary GM, Ivlev I, Houlihan ME, Yawn BP, Pasquale C, et al. (December 2021)."Developing a patient-driven chronic obstructive pulmonary disease (COPD) research agenda in the U.S."Journal of Patient-Reported Outcomes.5(1): 126.doi:10.1186/s41687-021-00399-7.PMC8643383.PMID34865193.
  236. ^Akers RM, Denbow DM (2008).Anatomy and Physiology of Domestic Animals.Wiley. p. 852.ISBN978-1-118-70115-7.
  237. ^Churg A, Wright JL (2007). "Animal models of cigarette smoke-induced chronic obstructive lung disease".Models of Exacerbations in Asthma and COPD.Contributions to Microbiology. Vol. 14. pp. 113–25.doi:10.1159/000107058.ISBN978-3-8055-8332-9.PMID17684336.
  238. ^"Recurrent Airway Obstruction in Horses - Respiratory System".Veterinary Manual.Retrieved7 July2021.
  239. ^Miller MS, Tilley LP, Smith FW (January 1989). "Cardiopulmonary disease in the geriatric dog and cat".The Veterinary Clinics of North America. Small Animal Practice.19(1): 87–102.doi:10.1016/S0195-5616(89)50007-X.PMID2646821.

Works cited

[edit]
[edit]