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

Stable COPD: Follow-up pharmacologic management

Stable COPD: Follow-up pharmacologic management
Literature review current through: Jan 2024.
This topic last updated: Nov 06, 2023.

INTRODUCTION — Chronic obstructive pulmonary disease (COPD) is a progressive obstructive lung disease with high morbidity and mortality. Current estimates suggest that approximately 10 percent of individuals aged 40 years or older have COPD, although the prevalence varies between countries and increases with age [1-3]. COPD is one of the leading causes of death among adults in the United States, resulting in over 140,000 deaths annually [4,5]. Prior to the onset of the COVID-19 (coronavirus disease 2019) pandemic, COPD was the third leading cause of death worldwide [6].

For the purposes of UpToDate topics, stable COPD refers to COPD in patients who are not currently experiencing an exacerbation of their disease. However, these patients may have experienced recent exacerbations with subsequent recovery to a new baseline or have ongoing COPD symptoms of varying intensity. Management of stable COPD typically occurs in the outpatient setting.

The pharmacologic management of patients with stable COPD who return in follow-up will be reviewed here. The clinical manifestations, diagnosis, comorbidities, and prognosis of COPD, as well as the management of refractory COPD, acute exacerbations, and alpha-1 antitrypsin deficiency are discussed separately.

(See "Chronic obstructive pulmonary disease: Diagnosis and staging".)

(See "Chronic obstructive pulmonary disease: Prognostic factors and comorbid conditions".)

(See "Stable COPD: Overview of management".)

(See "Stable COPD: Initial pharmacologic management".)

(See "Management of refractory chronic obstructive pulmonary disease".)

(See "COPD exacerbations: Management".)

(See "Treatment of alpha-1 antitrypsin deficiency".)

DISEASE METRICS — The Global Initiative Chronic Obstructive Lung Disease (GOLD) report uses the core metrics of dyspnea severity and frequency of exacerbations to categorize patients for pharmacologic management [7]. Initial management uses these metrics to divide patients into GOLD "ABE" groups for choice of initial therapy (algorithm 1). (See "Stable COPD: Initial pharmacologic management", section on 'Symptoms and risk of exacerbations'.)

Follow-up management for COPD uses the same core metrics of dyspnea and exacerbation history but differs from initial management in that it does not use GOLD groups for stratification. Importantly, follow-up management takes a similar approach to patients who have experienced exacerbations on therapy regardless of the degree of dyspnea.

ASSESSMENT

Symptoms and signs — For patients who return for follow-up management of COPD, we evaluate dyspnea and exercise tolerance using a validated instrument, such as the modified Medical Research Council (mMRC) dyspnea scale (calculator 1) or the COPD Assessment Test (CAT) (calculator 2) [7-10].

Physical examination focuses on changes associated with worsening airway obstruction and hyperinflation (hyperresonance, decreased breath sounds and diaphragmatic excursion, increased AP chest diameter, and wheezing). (See "Chronic obstructive pulmonary disease: Diagnosis and staging", section on 'Physical examination'.)

We also monitor additional symptoms associated with COPD (eg, cough, sputum, activity limitations, sleep disturbance), medication use, lung function, and oxygenation to ascertain whether an adequate response to therapy has been achieved and whether complications or comorbidities have developed, as described separately. (See "Stable COPD: Overview of management", section on 'Monitoring' and "Chronic obstructive pulmonary disease: Diagnosis and staging", section on 'Differential diagnosis' and "Chronic obstructive pulmonary disease: Prognostic factors and comorbid conditions".)

Exacerbation history — We support risk stratification for follow-up COPD therapy based on the occurrence of and severity of COPD exacerbations requiring treatment in the previous year (table 1). As with other COPD manifestations, exacerbation history requires continuous reassessment. At each follow-up visit, we review both the frequency and severity of COPD exacerbations and hospitalizations since the last visit, as well as the last adjustments of pharmacologic therapy. This is especially important as patients often obtain healthcare for exacerbations from different healthcare providers and care centers. The proposed Rome severity criteria for exacerbations (figure 1) are more objective than previous classification schemes that depended on treatment choices but can be more difficult to determine afterwards without access to detailed medical records.

The Global Initiative Chronic Obstructive Lung Disease (GOLD) strategy have been less specific regarding the threshold number and severity of exacerbations required to qualify for changes in management due to exacerbations during follow-up. Nevertheless, GOLD cites evidence favoring ICS use during follow-up care in COPD patients with one moderate exacerbation in the prior year and strongly favor ICS use in patients with one COPD exacerbation leading to hospitalization or two moderate exacerbations in the prior year [7].

Long-term studies evaluating therapeutic interventions for patients at risk for exacerbations have focused on patients at greatest exacerbation risk with one hospitalization or two or more exacerbations requiring systemic antibiotics or glucocorticoids as criteria to define a need and show a benefit of escalating COPD therapy [11,12]. However, accumulating evidence suggests that a single exacerbation requiring systemic therapies in a patient on long-acting maintenance treatment increases the subsequent risk of exacerbations over the next 12 to 36 months by 1.5- to 3-fold [13-17].

Additional risk factors for future exacerbations are discussed elsewhere. (See "COPD exacerbations: Clinical manifestations and evaluation", section on 'Risk factors'.)

Inhaler technique and adherence — Suboptimal inhaler technique and/or adherence are potential (and common) causes of a failed treatment response and should be recurrently explored with every patient. Device education and re-education are essential to successful follow-up care and should always be considered before adding or changing therapy. (See "Stable COPD: Overview of management", section on 'Inhaler technique'.)

A prospective study followed patients after hospitalization for COPD exacerbations and found suboptimal adherence to technique and on-time use in two-thirds [18]. Similarly, a systematic review of studies on inhaler technique found that on average two-thirds of patients with asthma or COPD made one or more errors in device use [19]. These studies support the need for regimen simplicity and re-enforcement of inhaler technique. (See "Patient education: Inhaler techniques in adults (Beyond the Basics)" and "The use of inhaler devices in adults".)

Review of smoking, comorbidities, and vaccination — Each follow-up visit is an opportunity to revisit smoking cessation, avoidance of exposure to inhalational particulates, fumes, or gases, vaccination against respiratory infections, contribution from potential comorbidities, and identification of patients for supplemental oxygen, pulmonary rehabilitation, and potential lung cancer screening. (See "Stable COPD: Overview of management" and "Screening for lung cancer".)

Blood eosinophils, inhaled corticosteroids, and exacerbations — Although the addition of inhaled corticosteroids (ICS) to pharmacologic regimens has reduced exacerbation risk in many trials, ICS are also associated with side effects, including the development of dysphonia, candidiasis, and a small increased frequency of lung infections. When deciding whether to add or taper ICS, evidence suggests that obtaining a peripheral blood eosinophil count may help guide therapy [7,11,20-26]. (See "Stable COPD: Initial pharmacologic management", section on 'Assessing disease pattern and severity' and "Role of inhaled glucocorticoid therapy in stable COPD" and "Major side effects of inhaled glucocorticoids".)

In general, higher levels of blood eosinophils in patients with exacerbations predict response to ICS therapy. We agree with the approach endorsed by GOLD, which advises using the following thresholds as an indication for advisability of ICS therapy in patients with exacerbations:

Patients with exacerbations and ≥300 eosinophils/microL are likely to have a favorable response to ICS and may have exacerbations following cessation of ICS therapy.

Patients with exacerbations and ≥100 but <300 eosinophils/microL may have a favorable response to ICS therapy.

Patients with exacerbations and <100 eosinophils/microL are unlikely to respond to ICS treatment and are at increased risk of infectious complications. ICS are not favored unless there is an alternative indication (eg, concomitant asthma).

When used as a biomarker to guide COPD therapy, blood eosinophil counts should be assessed during a period of disease stability and not during a course of systemic glucocorticoids or acute infection [27].

As an example of exacerbation risk associated with ICS use and eosinophil levels, a post hoc analysis of the Withdrawal of Inhaled Steroids during Optimized Bronchodilator Management (WISDOM) trial showed that among COPD patients with ≥1 exacerbation in the prior year, a blood eosinophil count ≥300 cells/microL identified patients at higher risk for exacerbations with discontinuation of steroids [25]. Among patients with ≥2 exacerbations per year, a blood eosinophil count ≥300 cells/microL was associated with roughly twice as many exacerbations after ICS discontinuation [26].

PHARMACOLOGIC ADJUSTMENT BASED ON ASSESSMENT — We agree with the Global Initiative Chronic Obstructive Lung Disease (GOLD) report strategy, which supports using quantified dyspnea and exacerbation frequency as the major metrics for adjustment of pharmacologic therapy in COPD (table 1) [7]. (See 'Disease metrics' above.)

Dyspnea well controlled and no exacerbations in the past year — In general, patients with good symptom control (ie, modified Medical Research Council [mMRC] grade <2 (calculator 1) or COPD Assessment Test [CAT] score <10 (calculator 2)) and no recent exacerbations should continue their current therapy [7]. As an exception, if current therapy includes inhaled glucocorticoids (inhaled corticosteroids [ICS]), tapering or discontinuing ICS is reasonable, particularly for those who did not benefit from the addition of ICS, have a low eosinophil count (<100 cells/microL), or are experiencing adverse events related to ICS therapy. If ICS are withdrawn, close patient monitoring is required. Any worsening in symptoms, activity intolerance, or oxygenation following treatment reduction should lead to a return to the prior maintenance therapy. (See 'Blood eosinophils, inhaled corticosteroids, and exacerbations' above and "Role of inhaled glucocorticoid therapy in stable COPD", section on 'Exacerbations'.)

Trials examining the effect of ICS withdrawal include the following:

For patients with good control of symptoms and <2 exacerbations/year on triple therapy, a real-world study in 914 patients suggests that ICS can be safely withdrawn without significant risk of health status decline (CAT score) or exacerbations [28]. One concern about the broad applicability of this study is that the original indications for ICS therapy were unclear.

In a 26-week trial, 527 patients with COPD on long-term triple therapy but infrequent exacerbations were randomly assigned to continue ICS (with tiotropium-salmeterol-fluticasone) or switch to dual therapy (glycopyrrolate-indacaterol) [29]. In the group without ICS, a small decrease was noted in forced expiratory volume in one second (FEV1), but there was no increase in the annualized rate of moderate or severe exacerbations. Patients with blood eosinophils ≥300/microL at baseline experienced a greater decline in FEV1 and a higher exacerbation risk.

Persistent dyspnea but no exacerbations in the past year

General approach to patients with dyspnea — For patients with persistent dyspnea or exercise limitation (ie, mMRC grade ≥2 (calculator 1) or CAT score ≥10 (calculator 2)) and no exacerbations in the past year, we generally agree with the pharmacologic approach recommended by the GOLD report (table 1) [7]. The mainstay of therapy for this group of patients is the use of dual bronchodilator (long-acting beta-agonist [LABA] and long-acting muscarinic-antagonist [LAMA]) therapy. ICS should be continued if it has been helping to reduce exacerbations and has been otherwise well tolerated. Patients with eosinophilia or possible asthma-COPD overlap (ACO) are also more likely to benefit from the use of ICS. Specific adjustments based on current regimens are detailed in the sections below.

Dyspnea on LAMA or LABA — For patients with COPD on a single long-acting bronchodilator experiencing ongoing dyspnea without exacerbations, we recommend use of a LAMA and LABA dual bronchodilator (available as a combination in a single inhaler).

A systematic review and meta-analysis of 10 trials (10,894 participants) compared combination LAMA (tiotropium) plus LABA (salmeterol, formoterol, or indacaterol) to tiotropium alone and found slightly better quality-of-life and a small increase in the postbronchodilator FEV1 with the combination [30].

Another systematic review and meta-analysis of 10 studies (9609 participants) found that LAMA/LABA (umeclidinium-vilanterol) combination therapy improved lung function compared with umeclidinium (LAMA), vilanterol (LABA), tiotropium (LABA), and fluticasone-salmeterol (LABA-ICS; mean improvement in trough FEV1 60, 110, 90, and 90 mL, respectively) [31]. Umeclidinium-vilanterol combination therapy also demonstrated a greater likelihood of a clinically significant improvement in dyspnea compared with either single-agent umeclidinium (56 versus 49 percent; risk ratio [RR] 1.14, 95% CI 1.05-1.24) or vilanterol (58 versus 48 percent; RR 1.21, 95% CI 1.12-1.31).

Dyspnea on LABA-ICS — LABA-ICS therapy is no longer recommended for most patients as initial treatment for COPD [7]. However, many patients use this combination based on its wide availability and endorsement in previous guideline recommendations. For patients without exacerbations but experiencing dyspnea on a LABA-ICS, we suggest addition of LAMA therapy either with or without continuation of the ICS. A recent history of frequent exacerbations that has improved with addition of ICS or a diagnosis of COPD and asthma (aka, COPD-A or ACO) argue in favor of changing LABA-ICS to LABA-LAMA-ICS triple therapy. Alternatively, if the initial indication for ICS is unclear, there has been no response to ICS treatment, or there have been significant ICS side effects, LABA-ICS can be changed to LAMA-LABA therapy.

Close monitoring is required for patients withdrawing from ICS to minimize any potential deterioration. Patients with blood eosinophils ≥300 cells/microL are more likely to experience an exacerbation after ICS withdrawal. (See 'Blood eosinophils, inhaled corticosteroids, and exacerbations' above.)

Very few studies focus on this group of patients, so this strategy is based primarily on evidence of beneficial addition of LAMA for dyspnea in patients on single bronchodilator therapy or improvement in dyspnea measures in trials examining patients with frequent exacerbations. (See 'Persistent exacerbations with or without dyspnea' below.)

KRONOS trial – The KRONOS trial compared the use of LABA-LAMA-ICS (budesonide-glycopyrrolate-formoterol 320/18/9.6 mcg) with LABA-LAMA therapy (glycopyrrolate-formoterol 18/9.6 mcg) and LABA-ICS (budesonide-formoterol 320/9.6 mcg or 400/12 mcg) in COPD patients who were symptomatic on two inhaled therapies [32]. Seventy-five percent of these patients had not had exacerbations within the past year. The groups using LAMA had much larger improvements in air movement (various FEV1-related measures) than patients on LABA-ICS. For example, change in predose trough FEV1 compared with baseline was 137 mL with LABA-LAMA-ICS, 110 mL with LABA-LAMA therapy, and 63 and 80 mL on the LABA-ICS combinations. Peak change in FEV1 postdosing was 381 mL with LABA-LAMA-ICS, 364 mL with LABA-LAMA therapy, and 275 or 291 mL with LABA-ICS. The small improvement with LABA-LAMA-ICS compared with LAMA-LABA was driven by patients with blood eosinophil counts >150 cells/microL. There were no consistent effects on dyspnea indices or rescue medication use. Regimens containing ICS reduced estimated exacerbation rates (approximately 0.5 exacerbations per year compared with 0.95 per year).

Other trials – Results from other studies of LABA-LAMA-ICS were all performed in patients with frequent exacerbations. They show a similar moderate to large improvement in predose FEV1 compared with LABA-ICS (63 to 170 mL), as well as modest improvements in symptomatic dyspnea (6 to 11 percent absolute increase in the number of patients reporting a clinically significant improvement in dyspnea by St. George's respiratory questionnaire [SGRQ]) [12,33-36]. Trials that compared LABA-LAMA-ICS with LABA-LAMA showed a much smaller change in spirometry measures (eg, 30 to 40 mL difference in trough FEV1) but similar improvements in respiratory symptoms by SGRQ, as well as fewer exacerbations [12,35-37]. (See 'Persistent exacerbations with or without dyspnea' below.)

Dyspnea on LAMA-LABA — For patients with low risk for exacerbations but significant dyspnea despite dual bronchodilator therapy, additional therapeutic options depend to some extent on the likelihood of benefit from the addition of ICS therapy.

Initial measures – We often initially attempt to switch dual bronchodilator therapy to an alternative inhaler device (eg, dry powder inhaler versus soft mist inhaler) or agents (eg, tiotropium-olodaterol versus umeclidinium-vilanterol). Additional work-up directed at other potential causes of dyspnea and evaluation for pulmonary rehabilitation are appropriate. (See "Pulmonary rehabilitation".)

For patients who continue to have dyspnea despite these interventions, our approach is as follows:

Patients with asthma-COPD overlap– Patients with clinical or spirometric features of asthma and COPD (variability of symptoms, history of atopy/allergies, strong bronchodilator response) may benefit from the addition of ICS therapy. (See "Asthma and COPD overlap (ACO)".)

Patients with peripheral eosinophilia – Trials comparing LABA-LAMA and LABA-LAMA-ICS suggest that ICS is most likely to improve lung function in patients with blood eosinophilia (≥100 to ≥150 cells/microL) [32,35]. We generally perform a trial of LAMA-LABA-ICS in patients with ≥100 eosinophils/microL before moving to other interventions. Invasive approaches (eg, bronchoscopic or surgical lung volume reduction or lung transplant) should be discussed with patients who meet criteria for these interventions. (See "Management of refractory chronic obstructive pulmonary disease", section on 'Lung Volume Reduction, in select patients with dyspnea' and "Management of refractory chronic obstructive pulmonary disease", section on 'Lung transplantation'.)

Patients without asthma-COPD overlap or eosinophilia – In patients with <100 eosinophils/microL and without asthma-COPD overlap features, either a switch to LAMA-LABA-ICS or the addition of low-dose theophylline can be attempted; however, these interventions usually do not have a major effect. Invasive approaches (eg, bronchoscopic or surgical lung volume reduction or lung transplant) should be discussed with patients who meet criteria for these interventions. (See "Management of refractory chronic obstructive pulmonary disease".)

Dyspnea on LAMA-LABA-ICS — For patients with dyspnea on triple therapy, other potential causes of dyspnea should be explored. Pulmonary rehabilitation often helps reduce dyspnea in this setting, particularly in more sedentary patients. Invasive approaches (eg, bronchoscopic or surgical lung volume reduction or lung transplant) may be of benefit in carefully selected patients. (See "Pulmonary rehabilitation" and "Management of refractory chronic obstructive pulmonary disease".)

Patients using LAMA-LABA-ICS who have not benefited from or have experienced adverse effects due to ICS should consider changing to LAMA-LABA combination therapy. ICS withdrawal requires close patient monitoring to avoid potential deterioration, particularly in patients who have elevated peripheral blood eosinophils ≥300 cells/microL. (See 'Blood eosinophils, inhaled corticosteroids, and exacerbations' above.)

Persistent exacerbations with or without dyspnea

General approach to patients with exacerbations — For patients with one or more exacerbations in the past year on their current regimen with or without associated persistent dyspnea, our approach is similar to the strategies outlined in the GOLD report [7]. We generally ensure these patients are on a baseline of LABA-LAMA therapy, as there are benefits in terms of dyspnea and possible benefits in exacerbation risk with only rare adverse effects. For most patients with exacerbations on this regimen, we then recommend a trial of ICS to reduce exacerbation risk; lack of efficacy or development of adverse effects from ICS should merit discontinuation. However, for patients with very low eosinophil counts (<100/microL) or persistent exacerbations despite triple therapy, we suggest either azithromycin maintenance therapy or roflumilast to reduce exacerbation risk. The decision between these agents is individualized based on their side effect profiles and patient comorbidities. (See "Management of refractory chronic obstructive pulmonary disease", section on 'Phosphodiesterase-4 inhibitors (Roflumilast)' and "Management of refractory chronic obstructive pulmonary disease", section on 'Macrolides and other chronic antibiotic therapy'.)

Specific pharmacologic adjustments based on current therapies are outlined below:

Exacerbations on LAMA or LABA — For patients with exacerbations on a LAMA or LABA alone and peripheral eosinophils ≤300 cells/microL, GOLD suggests a change to LAMA-LABA prior to instituting LAMA-LABA-ICS therapy, as dual bronchodilator therapy reduces dyspnea and may reduce exacerbations compared with single bronchodilator therapy [7,30,31]. However, the evidence for a reduction in exacerbations when a LABA is added to a LAMA is not consistent between trials [30,31,38]. (See "Stable COPD: Initial pharmacologic management", section on 'Use of dual bronchodilator therapy'.)

For patients with frequent exacerbations and a blood eosinophil count ≥300 cells/microL, we support increasing therapy to a LAMA-LABA-ICS, as advised by GOLD [7]. These patients have been found more likely to respond to ICS (LABA-ICS or LAMA-LABA-ICS) and are likely the main beneficiaries of the mortality gains seen with ICS therapy [23,39-41]. We also escalate to triple therapy in patients with severe exacerbations who require hospitalization, as these patients are at the highest risk for future COPD-associated hospitalizations and mortality. (See 'Blood eosinophils, inhaled corticosteroids, and exacerbations' above and 'Exacerbations on LAMA-LABA therapy' below and "Asthma and COPD overlap (ACO)" and "Stable COPD: Initial pharmacologic management", section on 'Group E patients presenting with elevated eosinophil counts or hospitalization'.)

Although multiple studies have shown the benefit of dual therapy with LABA-ICS versus monotherapy with LABA, we only use LABA-ICS in patients who are unable to take a LAMA or have asthma and COPD. Our preference for dual bronchodilator therapy is based largely on the low rate of adverse effects with LAMA therapy compared with ICS as well as the evidence in favor of triple therapy LABA-LAMA-ICS over LABA-ICS. The GOLD recommendations also endorse this approach [7]. LABA-ICS is an alternative in patients with eosinophil counts ≥100 cells/microL who do not tolerate LAMA therapy (table 1).

Exacerbations on LAMA-LABA therapy — For most patients with exacerbations despite dual therapy with LAMA-LABA, we suggest a change to LAMA-LABA-ICS, which is likely to reduce the frequency of exacerbations, improve lung function, and possibly improve mortality. Patients with higher levels of blood eosinophils (≥300 cells/microL) may experience the greatest likelihood of benefit, while those with very low baseline blood eosinophilia (<100 cells/microL) are least likely to benefit. Patients without eosinophilic inflammation may benefit more from alternative pharmacologic therapy with azithromycin and roflumilast to reduce their exacerbation risk. (See "Management of refractory chronic obstructive pulmonary disease", section on 'Macrolides and other chronic antibiotic therapy' and "Management of refractory chronic obstructive pulmonary disease", section on 'Phosphodiesterase-4 inhibitors (Roflumilast)'.)

The primary rationale for conversion of patients on LAMA-LABA to LAMA-LABA-ICS triple therapy is to achieve an improvement in lung function and decrease exacerbations. Supporting data come from several large clinical trials, which are reviewed in more detail below:

IMPACT – This trial compared a single, once-daily inhaler containing three agents (fluticasone furoate-umeclidinium-vilanterol) with a once-daily LAMA-LABA inhaler (umeclidinium-vilanterol) or a once-daily glucocorticoid-LABA inhaler (fluticasone furoate-vilanterol) in 10,355 patients with COPD who were at increased risk of exacerbations due to a combination of disease severity and exacerbation history [11]. The rate of moderate-to-severe exacerbations was lower with the triple inhaler compared with umeclidinium-vilanterol (annual rate of exacerbations 0.91 versus 1.21; RR 0.75, 95% CI 0.70-0.81). Additionally, lung function was better with triple therapy and hospitalizations were less frequent. However, the incidence of pneumonia was higher with triple therapy (95.8/1000 patient years) than with umeclidinium-vilanterol (61.2/1000 patient years; time-to-event hazard ratio [HR] 1.53, 95% CI 1.22-1.92). Questions have been raised about the role of withdrawal of ICS just prior to study onset as a factor in the findings from IMPACT [42], although this concept has also been contested.

Post hoc analysis suggests that the impact on exacerbation reduction, as well as lung function and dyspnea measures, was driven by patients with peripheral blood eosinophils >100/microL [43].

ETHOS – This trial compared a twice-daily inhaler containing three agents (budesonide-glycopyrrolate-formoterol) at two different ICS doses (320 mcg and 160 mcg budesonide) with a twice-daily LAMA-LABA inhaler (glycopyrrolate-formoterol) or a twice-daily ICS-LABA inhaler (budesonide-formoterol) in 8509 patients with COPD who were at increased risk for exacerbations [12]. The rate of moderate-to-severe exacerbations was nearly identically lower with either dose of the triple inhaler compared with LAMA-LABA (glycopyrrolate-formoterol; annual rate of exacerbations for the 320 mcg budesonide dose was 1.08 versus 1.42; RR 0.76, 95% CI 0.69-0.83). The incidence of pneumonia was higher with triple therapy (53.3/1000 patient years) than with glycopyrrolate-formoterol (37.3/1000 patient years).

TRIBUTE – This trial compared the single-inhaler triple therapy beclomethasone-formoterol-glycopyrrolate with glycopyrrolate-indacaterol in 1532 patients with severe to very severe COPD and increased exacerbation risk [37]. Over 52 weeks, triple therapy reduced exacerbation rates by approximately 15 percent (0.50 versus 0.59 exacerbations/patient per year for triple therapy and dual therapy, respectively). The effect was most pronounced in patients with blood eosinophil counts ≥2 percent. Overall adverse events were similar between the two study groups.

In addition to the impact of triple therapy on exacerbation rates, these trials demonstrated an improvement in mortality in high-risk patients with triple therapy compared with dual therapies. Although the magnitude of benefit appears small, the effect of these changes over time could become an important indication for LAMA-LABA-ICS therapy in the future. Meta-analysis of multiple trials has increased confidence in this result. (See "Role of inhaled glucocorticoid therapy in stable COPD", section on 'Exacerbations' and "Role of inhaled glucocorticoid therapy in stable COPD", section on 'Mortality'.)

In a meta-analysis of eight trials (including those discussed above) and over 25,000 patients with moderate-severe COPD, those assigned to LAMA-LABA-ICS compared with LAMA-LABA experienced a modest one-year mortality benefit (1.4 percent versus 2 percent, respectively, odds ratio 0.72, 95% CI 0.57-0.89) [39]. Subgroup analyses suggest that this benefit is likely driven by patients with ≥2 exacerbations per year and higher levels of blood eosinophils (≥200 cells / microL).

Post hoc analysis of mortality data from IMPACT demonstrated a likely benefit from triple therapy compared to LAMA-LABA (2.4 versus 3.2 percent one-year mortality, HR 0.72, 95% CI 0.53-0.99) [44]. Similar one-year mortality results were seen in ETHOS, although only improvements with standard-dose budesonide were statistically significant (high-dose budesonide-containing triple therapy versus. LAMA-LABA 1.4 versus 2.6 percent, HR 0.51, 95% CI 0.33-0.80; low-dose budesonide-containing triple therapy versus LAMA-LABA 2.1 versus 2.6 percent, HR 0.78, 95% CI 0.52-1.16) [45]. Other smaller trials, including TRIBUTE, were generally consistent with these findings.  

Exacerbations on LABA-ICS — LABA-ICS therapy is no longer recommended for most patients as initial treatment for COPD [7]. However, many patients use this combination based on its wide availability and endorsement in previous guideline recommendations. For COPD patients with recurrent exacerbations on LABA-ICS, we suggest step-up therapy to LAMA-LABA-ICS. These therapies have been directly compared in several randomized trials as outlined below.

For occasional patients with recurrent pneumonia, adverse side effects from ICS, or clear lack of response to ICS, switch to a LAMA-LABA is a reasonable alternative option. However, patients with a peripheral blood eosinophil count ≥300 cells/microL are more likely to experience an exacerbation if ICS therapy is discontinued. (See 'Blood eosinophils, inhaled corticosteroids, and exacerbations' above.)

The evidence for conversion of patients on LABA-ICS to LAMA-LABA-ICS triple therapy comes from several large clinical trials, which are reviewed in more detail below:

IMPACT and ETHOS – In the IMPACT trial, the rate of moderate-to-severe exacerbations was lower with the triple inhaler compared with fluticasone furoate-vilanterol (annual rate of exacerbations 0.91 versus 1.07; RR 0.85, 95% CI 0.80-0.90). There were also lung function and dyspnea improvements with triple therapy compared with LABA-ICS. In ETHOS, the change in annual rate of exacerbations was similar (1.08 versus 1.24; RR 0.87, 95% CI 0.79-0.95). These trials also evaluated triple therapy versus LAMA-LABA, as discussed in detail above. (See 'Dyspnea on LAMA or LABA' above.)

TRILOGY – This trial compared single-inhaler triple therapy with beclomethasone-glycopyrrolate-formoterol with beclomethasone-formoterol in 1368 patients with severe to very severe COPD and increased exacerbation risk [33]. Triple therapy reduced exacerbation rates by 23 percent (0.41 versus 0.53 annualized exacerbations; RR 0.77, 95% CI 0.65-0.92). The addition of the LAMA to the LABA-inhaled glucocorticoid increased trough FEV1 by 81 mL and reduced dyspnea. Overall, adverse events were similar between the two study arms.

FULFIL – This trial compared a once-daily single inhaler with fluticasone furoate-umeclidinium-vilanterol with twice-daily budesonide-formoterol in 1810 patients with moderate to severe COPD or with mild COPD at increased exacerbation risk [34]. At 26 weeks, triple therapy resulted in a mean increase in FEV1 (142 mL; 95% CI 126 to 158 mL), while dual therapy caused a decrease (-29 mL; 95% CI, -46 to -13 mL). A greater percentage of patients in the LABA-LAMA-ICS group had improvement in dyspnea compared with LABA-ICS (50 versus 41 percent). The annualized rate of moderate-to-severe exacerbations was reduced by 35 percent (0.22 versus 0.34; RR 0.65, 95% CI 0.49-0.86) with triple versus dual therapy.

A number of smaller trials and retrospective studies further support adding a LAMA to dual LABA-ICS therapy in patients with moderate to severe COPD [46-50].

Exacerbations on LAMA-LABA-ICS — Patients with COPD on LAMA-LABA-ICS who continue to have exacerbations should be considered to have refractory disease. Careful evaluation of inhaler technique is often a first step in managing these patients. Further pharmacologic management may include addition of a macrolide (ie, azithromycin) or roflumilast. Choice between these agents is often driven by drug interactions, patient comorbidities, and side effect profile. (See "Management of refractory chronic obstructive pulmonary disease", section on 'Phosphodiesterase-4 inhibitors (Roflumilast)' and "Management of refractory chronic obstructive pulmonary disease", section on 'Macrolides and other chronic antibiotic therapy'.)

Pulmonary rehabilitation should be considered in patients with persistent dyspnea or exacerbations despite triple therapy. Carefully selected patients may be candidates for lung volume reduction or lung transplant. These choices are discussed separately. (See "Management of refractory chronic obstructive pulmonary disease".)

REFRACTORY DISEASE — Some patients have severe COPD and are still symptomatic or continue to have exacerbations despite smoking cessation, an optimal regimen of inhaled medications (eg, long-acting muscarinic-antagonist/long-acting beta-agonist/inhaled corticosteroids [LAMA-LABA-ICS]), education about inhaler technique, pulmonary rehabilitation, and supplemental oxygen for severe hypoxemia. The management of these patients is discussed separately. (See "Management of refractory chronic obstructive pulmonary disease".)

FUTURE DIRECTIONS — Several novel therapies for COPD are being investigated.

Antieosinophilic biologics – The success of antieosinophilic biologics in severe eosinophilic asthma has led to investigational off-label use of these agents in COPD. Early data suggest modest effects in patients with elevated peripheral eosinophilia, but identification of the most effective agents and the subgroups of COPD patients who are most likely to benefit from these costly injectable therapies remains a challenge. (See "Treatment of severe asthma in adolescents and adults", section on 'Anti-IL-5 therapy' and "Treatment of severe asthma in adolescents and adults", section on 'Anti-lL-4 receptor alpha subunit antibody (dupilumab)'.)

DupilumabDupilumab is a monoclonal antibody that binds to the IL-4 receptor and is effective in reducing exacerbations and improving symptoms when used as add-on therapy in severe eosinophilic or allergic asthma. In patients with COPD, chronic bronchitis, elevated eosinophils (>300 cells/microL), and recurrent exacerbations despite optimized inhaled therapy, dupilumab reduced exacerbation rates and improved airway obstruction in one clinical trial. (See "Management of refractory chronic obstructive pulmonary disease", section on 'Frequent exacerbations despite azithromycin or roflumilast'.)

MepolizumabMepolizumab is a monoclonal antibody against interleukin (IL)-5 that is approved for use in eosinophilic asthma. In two parallel, randomized trials, 1138 patients with COPD, a blood eosinophil count ≥150 cells/microL, and moderate or severe exacerbations of COPD despite triple inhaler therapy were assigned to mepolizumab (100 mg in METREX, 100 or 300 mg in METREO) or placebo, administered subcutaneously every four weeks [51]. After one year, mepolizumab slightly reduced the rate of exacerbations compared with placebo (rate ratio 0.82, 95% CI 0.68-0.98 in METREX; RR 0.80, 95% CI 0.65-0.98 for mepolizumab 100 mg; and RR 0.86, 95% CI 0.70-1.05 for mepolizumab 300 mg in METREO).

BenralizumabBenralizumab is an IL-5 receptor alpha monoclonal antibody that is used as add-on therapy for patients with severe asthma and an eosinophilic phenotype. Mixed results have been reported with benralizumab in COPD [52]. In two parallel randomized trials, a total of 3910 patients with moderate to very severe COPD and a history of frequent exacerbations were assigned to take benralizumab (30 or 100 mg in GALATHEA; 10, 30, or 100 mg in TERRANOVA) or placebo subcutaneously every four weeks for two doses, then every eight weeks for a year [53]. None of the benralizumab doses achieved a significant reduction in annualized exacerbations relative to placebo among patients with a blood eosinophil count ≥220 cells/microL. In a subsequent post hoc subgroup analysis, patients with certain clinical characteristics (ie, ≥3 exacerbations in the previous year, on LAMA-LABA-ICS, and with blood eosinophil count ≥220 cells/microL) experienced a 30 percent reduction in exacerbations (RR 0.70, 95% CI 0.56-0.88) with benralizumab at the highest dose (100 mg every eight weeks) compared with placebo [54]. Further prospective study is needed to determine whether benralizumab would benefit this subgroup of COPD patients.

Ensifentrine – Ensifentrine is an inhaled selective dual phosphodiesterase-3 (PDE3) and phosphodiesterase-4 (PDE4) inhibitor that is a novel class of combined inhaled bronchodilator and anti-inflammatory agents. Acting through regulation of cyclic adenosine monophosphate (cAMP), PDE3 increases airway smooth muscle tone and PDE4 promotes inflammatory cell activation [55,56]. Dual inhibition of PDE3 and PDE4 has been suggested to enhance or have synergistic effects on both responses [57,58], prompting clinical development of a combined selective inhibitor [59,60].

In two concomitant phase 3 trials (ENHANCE-1 and ENHANCE-2), 760 and 789 patients with COPD, dyspnea (modified Medical Research Council [mMRC] scale ≥2 (calculator 1)), and moderate-severe airway obstruction (postbronchodilator forced expiratory volume in one second [FEV1] 30 to 70 percent predicted) received ensifentrine 3 mg or placebo twice daily via nebulizer [61]. Patients were receiving either no maintenance therapy, LABA±ICS, or LAMA±ICS at baseline, with these baseline therapies continued throughout the trials. No enrolled patients were receiving either dual LAMA-LABA or LAMA-LABA-ICS therapy. Ensifentrine demonstrated efficacy as a bronchodilator, leading to significant improvements in peak and trough FEV1. Patients using ensifentrine experienced potentially meaningful improvements in dyspnea (1.0- and 0.9-point increase in the Transition Dyspnea Index compared with placebo). Ensifentrine also improved rates of exacerbations requiring systemic corticosteroids or antibiotics in both trials (rate ratio 0.64, 95% CI 0.4-1.0 and rate ratio 0.57, 95% CI 0.4-0.9) irrespective of patient baseline therapy. Adverse events, including gastrointestinal side effects often seen with oral PDE4 inhibitors, were minimal and similar to placebo.

The prospect of a novel well-tolerated and effective inhaled bronchodilator/anti-inflammatory agent is welcome and may lead to additional therapeutic options for patients with COPD. After regulatory approval, additional study will be needed to determine the optimal strategy for use of this therapy in COPD management.

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

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

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

Basics topics (see "Patient education: Pulmonary rehabilitation (The Basics)")

SUMMARY AND RECOMMENDATIONS

Disease metrics – At follow-up visits for COPD, we focus our evaluation on symptoms (dyspnea and exercise tolerance) and on exacerbation history. The Global Initiative Chronic Obstructive Lung Disease (GOLD) “ABE” groups that are used to guide initial therapy are not used for follow-up adjustments of medication. (See 'Disease metrics' above.)

Assessment – Dyspnea should be determined using validated instruments. Physical examination may assist in evaluation of worsening hyperinflation and airflow obstruction. Exacerbation history and severity in the prior year should also be reassessed. We concomitantly evaluate inhaler technique and adherence, review tobacco use and comorbidities, administer vaccines against respiratory pathogens, and measure blood eosinophil levels in selected patients. (See 'Assessment' above.)

Pharmacologic adjustment – For patients who have persistent symptoms or recurrent exacerbations, pharmacologic therapy should be adjusted based on the patient’s current medications (table 1). Other causes of dyspnea and recurrent exacerbations should be explored and treated as indicated. (See "Stable COPD: Initial pharmacologic management".)

Patients without dyspnea or exacerbations – Patients with good symptom control (ie, modified Medical Research Council [mMRC] grade 0 or 1 (calculator 1) or COPD Assessment Test [CAT] score <10 (calculator 2)) and low exacerbation risk should continue their current therapy. As an exception, if current therapy includes inhaled glucocorticoid (inhaled corticosteroid [ICS]), tapering or discontinuing the ICS is often well tolerated, particularly in those with low blood eosinophil counts. (See 'Dyspnea well controlled and no exacerbations in the past year' above.)

Patients with persistent dyspnea but without exacerbations – The mainstay of therapy for COPD patients with persistent dyspnea (ie, mMRC grade ≥2 (calculator 1) or CAT score ≥10 (calculator 2)) is the use of dual bronchodilator (long-acting beta-agonist/long-acting muscarinic-antagonist [LABA-LAMA]) therapy. ICS should be continued if it has been helping reduce exacerbations and has otherwise been well tolerated. (See 'General approach to patients with dyspnea' above.)

Therapy can be adjusted step-wise, as follows:

-For patients on monotherapy with a LAMA or LABA with dyspnea, we suggest changing to dual LAMA-LABA therapy rather than switching to an alternative single agent (Grade 2B). (See 'Dyspnea on LAMA or LABA' above.)

-For patients previously using a LABA-ICS combination with dyspnea, changing to either a LAMA-LABA combination or to triple therapy (LAMA-LABA-ICS) is appropriate. Patients with eosinophilia or possible asthma-COPD overlap (ACO) are more likely to benefit from the use of ICS, while those with blood eosinophils <100 microL are reasonable candidates for dropping ICS. (See 'Dyspnea on LABA-ICS' above.)

-For patients on LAMA-LABA therapy, a different preparation or delivery system may be more effective. Those with eosinophils >100 cells/microL may benefit from a trial of LAMA-LABA-ICS therapy. In addition, for patients using a LAMA-LABA or LAMA-LABA-ICS inhaler, low-dose theophylline and/or nonpharmacologic options, such as pulmonary rehabilitation, bronchoscopic or surgical lung volume reduction, or lung transplantation, may be of benefit. (See 'Dyspnea on LAMA or LABA' above and 'Dyspnea on LAMA-LABA-ICS' above.)

Patients with exacerbations – For patients with one or more exacerbations in the past year with or without persistent dyspnea, our step-wise approach is as follows:

-For patients on LABA or LAMA monotherapy, we suggest initiation of dual bronchodilator therapy with a LABA-LAMA rather than an alternative single agent or initiation of ICS (Grade 2C). As an exception, patients with a blood eosinophil count ≥300 cells/microL or asthma and COPD may be more likely to respond to LAMA-LABA-ICS. (See 'Exacerbations on LAMA or LABA' above.)

-For patients using dual therapy (LAMA-LABA or LABA-ICS) and eosinophils ≥100/microL, we suggest changing to triple therapy (LAMA-LABA-ICS) rather than other dual therapy options (Grade 2B). (See 'Exacerbations on LAMA-LABA therapy' above and 'Exacerbations on LABA-ICS' above.)

-For patients on LAMA-LABA with eosinophils <100/microL or LAMA-LABA-ICS who continue to have exacerbations, additional treatment options include a macrolide (ie, azithromycin) or roflumilast, with the choice guided by patient comorbidities and tolerance of side effects. (See 'Exacerbations on LAMA-LABA-ICS' above and "Management of refractory chronic obstructive pulmonary disease", section on 'Macrolides and other chronic antibiotic therapy' and "Management of refractory chronic obstructive pulmonary disease", section on 'Phosphodiesterase-4 inhibitors (Roflumilast)'.)

  1. Ford ES, Croft JB, Mannino DM, et al. COPD surveillance--United States, 1999-2011. Chest 2013; 144:284.
  2. Burney P, Jithoo A, Kato B, et al. Chronic obstructive pulmonary disease mortality and prevalence: the associations with smoking and poverty--a BOLD analysis. Thorax 2014; 69:465.
  3. Ford ES, Mannino DM, Wheaton AG, et al. Trends in the prevalence of obstructive and restrictive lung function among adults in the United States: findings from the National Health and Nutrition Examination surveys from 1988-1994 to 2007-2010. Chest 2013; 143:1395.
  4. Kochanek KD, Murphy SL, Xu J, Arias E. Deaths: Final Data for 2017. National Vital Statistics Reports 2019. https://www.cdc.gov/nchs/data/nvsr/nvsr68/nvsr68_09-508.pdf (Accessed on October 09, 2021).
  5. Ahmad FB, Anderson RN. The Leading Causes of Death in the US for 2020. JAMA 2021; 325:1829.
  6. World Health Organization. The top 10 causes of death. https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death (Accessed on May 28, 2019).
  7. Global Initiative for Chronic Obstructive Lung Disease (GOLD). Global Strategy for the Diagnosis, Management and Prevention of Chronic Obstructive Pulmonary Disease: 2024 Report. www.goldcopd.org www.goldcopd.org (Accessed on November 16, 2023).
  8. Jones PW, Tabberer M, Chen WH. Creating scenarios of the impact of COPD and their relationship to COPD Assessment Test (CAT™) scores. BMC Pulm Med 2011; 11:42.
  9. COPD Assessment Test (CAT) for outside the United States. http://www.catestonline.org/ (Accessed on February 06, 2019).
  10. Gupta N, Pinto LM, Morogan A, Bourbeau J. The COPD assessment test: a systematic review. Eur Respir J 2014; 44:873.
  11. Lipson DA, Barnhart F, Brealey N, et al. Once-Daily Single-Inhaler Triple versus Dual Therapy in Patients with COPD. N Engl J Med 2018; 378:1671.
  12. Rabe KF, Martinez FJ, Ferguson GT, et al. Triple Inhaled Therapy at Two Glucocorticoid Doses in Moderate-to-Very-Severe COPD. N Engl J Med 2020; 383:35.
  13. Vogelmeier CF, Diesing J, Kossack N, et al. COPD Exacerbation History and Impact on Future Exacerbations - 8-Year Retrospective Observational Database Cohort Study from Germany. Int J Chron Obstruct Pulmon Dis 2021; 16:2407.
  14. Whittaker H, Rubino A, Müllerová H, et al. Frequency and Severity of Exacerbations of COPD Associated with Future Risk of Exacerbations and Mortality: A UK Routine Health Care Data Study. Int J Chron Obstruct Pulmon Dis 2022; 17:427.
  15. Marott JL, Çolak Y, Ingebrigtsen TS, et al. Exacerbation history, severity of dyspnoea and maintenance treatment predicts risk of future exacerbations in patients with COPD in the general population. Respir Med 2022; 192:106725.
  16. Haughney J, Lee AJ, Nath M, et al. The long-term clinical impact of COPD exacerbations: a 3-year observational study (SHERLOCK). Ther Adv Respir Dis 2022; 16:17534666211070139.
  17. Vanfleteren LEGW, Lindberg A, Zhou C, et al. Exacerbation Risk and Mortality in Global Initiative for Chronic Obstructive Lung Disease Group A and B Patients with and without Exacerbation History. Am J Respir Crit Care Med 2023; 208:163.
  18. Sulaiman I, Cushen B, Greene G, et al. Objective Assessment of Adherence to Inhalers by Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 195:1333.
  19. Sanchis J, Gich I, Pedersen S, Aerosol Drug Management Improvement Team (ADMIT). Systematic Review of Errors in Inhaler Use: Has Patient Technique Improved Over Time? Chest 2016; 150:394.
  20. Leung JM, Sin DD. Asthma-COPD overlap syndrome: pathogenesis, clinical features, and therapeutic targets. BMJ 2017; 358:j3772.
  21. Bafadhel M, McKenna S, Terry S, et al. Blood eosinophils to direct corticosteroid treatment of exacerbations of chronic obstructive pulmonary disease: a randomized placebo-controlled trial. Am J Respir Crit Care Med 2012; 186:48.
  22. Bafadhel M, McKenna S, Terry S, et al. Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers. Am J Respir Crit Care Med 2011; 184:662.
  23. Bafadhel M, Peterson S, De Blas MA, et al. Predictors of exacerbation risk and response to budesonide in patients with chronic obstructive pulmonary disease: a post-hoc analysis of three randomised trials. Lancet Respir Med 2018; 6:117.
  24. Pavord ID, Lettis S, Anzueto A, Barnes N. Blood eosinophil count and pneumonia risk in patients with chronic obstructive pulmonary disease: a patient-level meta-analysis. Lancet Respir Med 2016; 4:731.
  25. Watz H, Tetzlaff K, Wouters EF, et al. Blood eosinophil count and exacerbations in severe chronic obstructive pulmonary disease after withdrawal of inhaled corticosteroids: a post-hoc analysis of the WISDOM trial. Lancet Respir Med 2016; 4:390.
  26. Calverley PMA, Tetzlaff K, Vogelmeier C, et al. Eosinophilia, Frequent Exacerbations, and Steroid Response in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 196:1219.
  27. Echevarria C, Steer J, Prasad A, et al. Admission blood eosinophil count, inpatient death and death at 1 year in exacerbating patients with COPD. Thorax 2023; 78:1090.
  28. Rossi A, Guerriero M, Corrado A, OPTIMO/AIPO Study Group. Withdrawal of inhaled corticosteroids can be safe in COPD patients at low risk of exacerbation: a real-life study on the appropriateness of treatment in moderate COPD patients (OPTIMO). Respir Res 2014; 15:77.
  29. Chapman KR, Hurst JR, Frent SM, et al. Long-Term Triple Therapy De-escalation to Indacaterol/Glycopyrronium in Patients with Chronic Obstructive Pulmonary Disease (SUNSET): A Randomized, Double-Blind, Triple-Dummy Clinical Trial. Am J Respir Crit Care Med 2018; 198:329.
  30. Farne HA, Cates CJ. Long-acting beta2-agonist in addition to tiotropium versus either tiotropium or long-acting beta2-agonist alone for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2015; :CD008989.
  31. Rodrigo GJ, Neffen H. A Systematic Review of the Efficacy and Safety of a Fixed-Dose Combination of Umeclidinium and Vilanterol for the Treatment of COPD. Chest 2015; 148:397.
  32. Ferguson GT, Rabe KF, Martinez FJ, et al. Triple therapy with budesonide/glycopyrrolate/formoterol fumarate with co-suspension delivery technology versus dual therapies in chronic obstructive pulmonary disease (KRONOS): a double-blind, parallel-group, multicentre, phase 3 randomised controlled trial. Lancet Respir Med 2018; 6:747.
  33. Singh D, Papi A, Corradi M, et al. Single inhaler triple therapy versus inhaled corticosteroid plus long-acting β2-agonist therapy for chronic obstructive pulmonary disease (TRILOGY): a double-blind, parallel group, randomised controlled trial. Lancet 2016; 388:963.
  34. Lipson DA, Barnacle H, Birk R, et al. FULFIL Trial: Once-Daily Triple Therapy for Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2017; 196:438.
  35. Rabe KF, Martinez FJ, Singh D, et al. Improvements in lung function with budesonide/glycopyrrolate/formoterol fumarate metered dose inhaler versus dual therapies in patients with COPD: a sub-study of the ETHOS trial. Ther Adv Respir Dis 2021; 15:17534666211034329.
  36. Martinez FJ, Rabe KF, Ferguson GT, et al. Benefits of budesonide/glycopyrrolate/formoterol fumarate (BGF) on symptoms and quality of life in patients with COPD in the ETHOS trial. Respir Med 2021; 185:106509.
  37. Papi A, Vestbo J, Fabbri L, et al. Extrafine inhaled triple therapy versus dual bronchodilator therapy in chronic obstructive pulmonary disease (TRIBUTE): a double-blind, parallel group, randomised controlled trial. Lancet 2018; 391:1076.
  38. Calverley PMA, Anzueto AR, Carter K, et al. Tiotropium and olodaterol in the prevention of chronic obstructive pulmonary disease exacerbations (DYNAGITO): a double-blind, randomised, parallel-group, active-controlled trial. Lancet Respir Med 2018; 6:337.
  39. Chen H, Deng ZX, Sun J, et al. Association of Inhaled Corticosteroids With All-Cause Mortality Risk in Patients With COPD: A Meta-analysis of 60 Randomized Controlled Trials. Chest 2023; 163:100.
  40. Siddiqui SH, Guasconi A, Vestbo J, et al. Blood Eosinophils: A Biomarker of Response to Extrafine Beclomethasone/Formoterol in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2015; 192:523.
  41. Suissa S. Single-inhaler triple versus dual bronchodilator therapy for GOLD group E and other exacerbating patients with COPD: real-world comparative effectiveness and safety. Eur Respir J 2023; 62.
  42. Suissa S, Drazen JM. Making Sense of Triple Inhaled Therapy for COPD. N Engl J Med 2018; 378:1723.
  43. Pascoe S, Barnes N, Brusselle G, et al. Blood eosinophils and treatment response with triple and dual combination therapy in chronic obstructive pulmonary disease: analysis of the IMPACT trial. Lancet Respir Med 2019; 7:745.
  44. Lipson DA, Crim C, Criner GJ, et al. Reduction in All-Cause Mortality with Fluticasone Furoate/Umeclidinium/Vilanterol in Patients with Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2020; 201:1508.
  45. Martinez FJ, Rabe KF, Ferguson GT, et al. Reduced All-Cause Mortality in the ETHOS Trial of Budesonide/Glycopyrrolate/Formoterol for Chronic Obstructive Pulmonary Disease. A Randomized, Double-Blind, Multicenter, Parallel-Group Study. Am J Respir Crit Care Med 2021; 203:553.
  46. Tashkin DP, Celli B, Senn S, et al. A 4-year trial of tiotropium in chronic obstructive pulmonary disease. N Engl J Med 2008; 359:1543.
  47. Lee TA, Wilke C, Joo M, et al. Outcomes associated with tiotropium use in patients with chronic obstructive pulmonary disease. Arch Intern Med 2009; 169:1403.
  48. Short PM, Williamson PA, Elder DH, et al. The impact of tiotropium on mortality and exacerbations when added to inhaled corticosteroids and long-acting β-agonist therapy in COPD. Chest 2012; 141:81.
  49. Aaron SD, Vandemheen KL, Fergusson D, et al. Tiotropium in combination with placebo, salmeterol, or fluticasone-salmeterol for treatment of chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med 2007; 146:545.
  50. Karner C, Cates CJ. The effect of adding inhaled corticosteroids to tiotropium and long-acting beta(2)-agonists for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2011; :CD009039.
  51. Pavord ID, Chanez P, Criner GJ, et al. Mepolizumab for Eosinophilic Chronic Obstructive Pulmonary Disease. N Engl J Med 2017; 377:1613.
  52. Donovan T, Milan SJ, Wang R, et al. Anti-IL-5 therapies for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2020; 12:CD013432.
  53. Criner GJ, Celli BR, Brightling CE, et al. Benralizumab for the Prevention of COPD Exacerbations. N Engl J Med 2019; 381:1023.
  54. Criner GJ, Celli BR, Singh D, et al. Predicting response to benralizumab in chronic obstructive pulmonary disease: analyses of GALATHEA and TERRANOVA studies. Lancet Respir Med 2020; 8:158.
  55. Banner KH, Press NJ. Dual PDE3/4 inhibitors as therapeutic agents for chronic obstructive pulmonary disease. Br J Pharmacol 2009; 157:892.
  56. Martin C, Burgel PR, Roche N. Inhaled Dual Phosphodiesterase 3/4 Inhibitors for the Treatment of Patients with COPD: A Short Review. Int J Chron Obstruct Pulmon Dis 2021; 16:2363.
  57. Schmidt DT, Watson N, Dent G, et al. The effect of selective and non-selective phosphodiesterase inhibitors on allergen- and leukotriene C(4)-induced contractions in passively sensitized human airways. Br J Pharmacol 2000; 131:1607.
  58. Milara J, Navarro A, Almudéver P, et al. Oxidative stress-induced glucocorticoid resistance is prevented by dual PDE3/PDE4 inhibition in human alveolar macrophages. Clin Exp Allergy 2011; 41:535.
  59. Franciosi LG, Diamant Z, Banner KH, et al. Efficacy and safety of RPL554, a dual PDE3 and PDE4 inhibitor, in healthy volunteers and in patients with asthma or chronic obstructive pulmonary disease: findings from four clinical trials. Lancet Respir Med 2013; 1:714.
  60. Ferguson GT, Kerwin EM, Rheault T, et al. A Dose-Ranging Study of the Novel Inhaled Dual PDE 3 and 4 Inhibitor Ensifentrine in Patients with COPD Receiving Maintenance Tiotropium Therapy. Int J Chron Obstruct Pulmon Dis 2021; 16:1137.
  61. Anzueto A, Barjaktarevic IZ, Siler TM, et al. Ensifentrine, a Novel Phosphodiesterase 3 and 4 Inhibitor for the Treatment of Chronic Obstructive Pulmonary Disease: Randomized, Double-Blind, Placebo-controlled, Multicenter Phase III Trials (the ENHANCE Trials). Am J Respir Crit Care Med 2023; 208:406.
Topic 122160 Version 25.0

References

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