Skip to main content
The Cochrane Database of Systematic Reviews logoLink to The Cochrane Database of Systematic Reviews
. 2020 May 1;2020(5):CD002309. doi: 10.1002/14651858.CD002309.pub6

Phosphodiesterase‐4 inhibitors for chronic obstructive pulmonary disease

Sadia Janjua 1, Rebecca Fortescue 1, Phillippa Poole 2,
Editor: Cochrane Airways Group
PMCID: PMC7193764  PMID: 32356609

Abstract

Background

Chronic obstructive pulmonary disease (COPD) is associated with cough, sputum production or dyspnoea, and a reduction in lung function, quality of life, and life expectancy. Apart from smoking cessation, no other treatments that slow lung function decline are available. Roflumilast and cilomilast are oral phosphodiesterase‐4 (PDE₄) inhibitors proposed to reduce the airway inflammation and bronchoconstriction seen in COPD. This Cochrane Review was first published in 2011, and was updated in 2017 and 2020.

Objectives

To evaluate the efficacy and safety of oral PDE₄ inhibitors for management of stable COPD.

Search methods

We identified randomised controlled trials (RCTs) from the Cochrane Airways Trials Register (date of last search 9 March 2020). We found other trials at web‐based clinical trials registers.

Selection criteria

We included RCTs if they compared oral PDE₄ inhibitors with placebo in people with COPD. We allowed co‐administration of standard COPD therapy.

Data collection and analysis

We used standard Cochrane methods. Two independent review authors selected trials for inclusion, extracted data, and assessed risk of bias. We resolved discrepancies by involving a third review author. We assessed our confidence in the evidence by using GRADE recommendations. Primary outcomes were change in lung function (minimally important difference (MID) = 100 mL) and quality of life (scale 0 to 100; higher score indicates more limitations).

Main results

We found 42 RCTs that met the inclusion criteria and were included in the analyses for roflumilast (28 trials with 18,046 participants) or cilomilast (14 trials with 6457 participants) or tetomilast (1 trial with 84 participants), with a duration between six weeks and one year or longer. These trials included people across international study centres with moderate to very severe COPD (Global Initiative for Chronic Obstructive Lung Disease (GOLD) grades II to IV), with mean age of 64 years.

We judged risks of selection bias, performance bias, and attrition bias as low overall amongst the 39 published and unpublished trials.

Lung function

Treatment with a PDE₄ inhibitor was associated with a small, clinically insignificant improvement in forced expiratory volume in one second (FEV₁) over a mean of 40 weeks compared with placebo (mean difference (MD) 49.33 mL, 95% confidence interval (CI) 44.17 to 54.49; participants = 20,815; studies = 29; moderate‐certainty evidence). Forced vital capacity (FVC) and peak expiratory flow (PEF) were also improved over 40 weeks (FVC: MD 86.98 mL, 95% CI 74.65 to 99.31; participants = 22,108; studies = 17; high‐certainty evidence; PEF: MD 6.54 L/min, 95% CI 3.95 to 9.13; participants = 4245; studies = 6; low‐certainty evidence).

Quality of life

Trials reported improvements in quality of life over a mean of 33 weeks (St George's Respiratory Questionnaire (SGRQ) MD ‐1.06 units, 95% CI ‐1.68 to ‐0.43; participants = 7645 ; moderate‐certainty evidence).

Incidence of exacerbations

Treatment with a PDE₄ inhibitor was associated with a reduced likelihood of COPD exacerbation over a mean of 40 weeks (odds ratio (OR) 0.78, 95% CI 0.73 to 0.84; participants = 20,382; studies = 27; high‐certainty evidence), that is, for every 100 people treated with PDE₄ inhibitors, five more remained exacerbation‐free during the study period compared with those given placebo (number needed to treat for an additional beneficial outcome (NNTB) 20, 95% CI 16 to 27). No change in COPD‐related symptoms nor in exercise tolerance was found.

Adverse events

More participants in the treatment groups experienced an adverse effect compared with control participants over a mean of 39 weeks (OR 1.30, 95% CI 1.22 to 1.38; participants = 21,310; studies = 30; low‐certainty evidence). Participants experienced a range of gastrointestinal symptoms such as diarrhoea, nausea, vomiting, or dyspepsia. Diarrhoea was more commonly reported with PDE₄ inhibitor treatment (OR 3.20, 95% CI 2.74 to 3.50; participants = 20,623; studies = 29; high‐certainty evidence), that is, for every 100 people treated with PDE₄ inhibitors, seven more suffered from diarrhoea during the study period compared with those given placebo (number needed to treat for an additional harmful outcome (NNTH) 15, 95% CI 13 to 17). The likelihood of psychiatric adverse events was higher with roflumilast 500 µg than with placebo (OR 2.13, 95% CI 1.79 to 2.54; participants = 11,168; studies = 15 (COPD pool data); moderate‐certainty evidence). Roflumilast in particular was associated with weight loss during the trial period and with an increase in insomnia and depressive mood symptoms.

Participants treated with PDE₄ inhibitors were more likely to withdraw from trial participation; on average, 14% in the treatment groups withdrew compared with 8% in the control groups.

Mortality

No effect on mortality was found (OR 0.98, 95% CI 0.77 to 1.24; participants = 19,786; studies = 27; moderate‐certainty evidence), although mortality was a rare event during these trials.

Authors' conclusions

For this current update, five new studies from the 2020 search contributed to existing findings but made little impact on outcomes described in earlier versions of this review.

PDE₄ inhibitors offered a small benefit over placebo in improving lung function and reducing the likelihood of exacerbations in people with COPD; however, they had little impact on quality of life or on symptoms. Gastrointestinal adverse effects and weight loss were common, and the likelihood of psychiatric symptoms was higher, with roflumilast 500 µg.

The findings of this review provide cautious support for the use of PDE₄ inhibitors in COPD. In accordance with GOLD 2020 guidelines, they may have a place as add‐on therapy for a subgroup of people with persistent symptoms or exacerbations despite optimal COPD management (e.g. people whose condition is not controlled by fixed‐dose long‐acting beta₂‐agonist (LABA) and inhaled corticosteroid (ICS) combinations). More longer‐term trials are needed to determine whether or not PDE₄ inhibitors modify FEV₁ decline, hospitalisation, or mortality in COPD.

Plain language summary

Phosphodiesterase‐4 inhibitors for people with chronic obstructive pulmonary disease (COPD)

Background

COPD is a progressive lung condition caused by damage from harmful chemicals breathed in and is predominantly seen in people who smoke tobacco. These chemicals cause inflammation and lung damage and increase mucus production in the lungs. This leads to periods of breathlessness and coughing called exacerbations (or flare‐ups). Exacerbations make it harder for people to do their day‐to‐day tasks. Exacerbations become more frequent and severe over time. People vary in terms of how they are affected by COPD. This is related in part to the severity of the disease but also to differences in response to medicines, as well as fitness and co‐existent conditions. For most people, the only way to prevent further lung damage is to stop smoking.

Medicines prescribed to manage COPD generally aim to improve symptoms, reduce exacerbations, or both. In early stages, taking bronchodilators makes breathing easier by relaxing muscles in the lungs and widening airways, allowing more air to move freely into and out of the lungs.

Some long‐acting agents may reduce exacerbations. For example, steroid inhalers reduce inflammation in the lungs and thus modestly reduce the number of exacerbations.

Phosphodiesterase‐4 (PDE₄) inhibitors are a relatively new class of medicines marketed to improve COPD. They have both bronchodilator and anti‐inflammatory effects. Two currently available medicines ‐ roflumilast and cilomilast ‐ are taken as a tablet. We collated and analysed results of existing trials to define the benefits and risks of PDE₄ inhibitors in COPD.

Key results

Data analysis included 42 studies in 24,587 adults with moderate to very severe disease who discontinued other regular COPD medications. Some trials allowed people to carry on using their usual COPD medicines. Most trials were funded by manufacturers of PDE₄ inhibitors.

PDE₄ inhibitors provided a small benefit in improving lung function measurements (forced expiratory volume in one second (FEV₁), forced vital capacity (FVC), and peak expiratory flow (PEF)). PDE₄ inhibitors also reduced the likelihood of COPD‐related exacerbations. We found that 28 out of 100 people taking PDE₄ inhibitors every day for a year would experience at least one exacerbation, which was five fewer than for people who did not receive these medicines.

PDE₄ inhibitors provided a small benefit in reducing breathlessness and improving quality of life. Around 5% to 10% of people who received roflumilast or cilomilast reported side effects such as diarrhoea, nausea, and vomiting. We expected that 11 out of 100 people taking PDE₄ inhibitors every day for 39 weeks would experience an episode of diarrhoea, which was seven more than for those not receiving PDE₄ inhibitors. We found that 7 people out of 100 were likely to experience a psychiatric event with roflumilast 500 µg. A two‐ to three‐fold increase in risk of sleep or mood disturbance was found with roflumilast 500 μg, although overall the total number of reported incidents was low. There was no effect on death rates. Effects were the same regardless of the severity of COPD, or whether other medicines for COPD were being taken.

Quality of the evidence

We were moderately certain about data for lung function and quality of life. We were highly certain of evidence for side effects such as diarrhoea and of data for exacerbations.

Results seen in trials published in journals by pharmaceutical companies show greater benefit of these medicines than those that were unpublished. Psychiatric adverse effects data remain unpublished.

Conclusions

We support the use of PDE₄ inhibitors for COPD, but with caution. PDE₄ inhibitors provided a small benefit in improving lung function and reducing the likelihood of COPD exacerbations, but they had little impact on quality of life and COPD symptoms. Side effects including diarrhoea and weight loss were common.

PDE₄ inhibitors may be best used as add‐on therapy for a subgroup of people with persistent symptoms or exacerbations despite otherwise optimal COPD management (e.g. people whose condition was not controlled on fixed‐dose long‐acting beta₂‐agonist (LABA) and inhaled corticosteroid (ICS) combinations). This is in accordance with GOLD 2020 guidelines. Longer‐term trials are necessary to get a more accurate estimate of the benefits and safety of these medicines over time, including determining whether they slow COPD disease progression.

Summary of findings

Background

Description of the condition

Chronic obstructive pulmonary disease (COPD) is one of the leading causes of global morbidity and mortality, resulting in a growing social and economic burden (GOLD 2020). In 2002, COPD was estimated to be the fifth leading cause of death, responsible for approximately 4.8% of total deaths worldwide, and it is projected to rise to fourth position by the year 2030 (Mathers 2005).

COPD is an overarching term that includes two lung conditions: chronic bronchitis and emphysema. These lung conditions cause narrowing of the airways and overinflation of the alveoli, leading to difficulty in breathing. Diagnosis of COPD by the Global Initiative for Chronic Obstructive Lung Disease (GOLD) says that it is a "heterogeneous disease that is characterized by persistent respiratory symptoms and airflow limitation that is due to airway and/or alveolar abnormalities usually caused by exposure to noxious particles or gases and influenced by host factors including abnormal lung development" (GOLD 2020). COPD may be punctuated by periods of acute worsening of respiratory symptoms, called exacerbations. Besides exposures, host factors predispose individuals to develop COPD. Comorbidities contribute to overall severity and mortality in individual people (GOLD 2020). Diagnosis is based on a history of exposure to risk factors for this disease and symptoms of cough and sputum production or dyspnoea (shortness of breath). Spirometry is required for diagnosis, with airflow obstruction confirmed by a post‐bronchodilator forced expiratory volume in one second (FEV₁)/forced vital capacity (FVC) of 0.7 or lower (Celli 2004). Life expectancy is reduced among people diagnosed with COPD, and although prognosis is variable, age and FEV₁ are the strongest predictors of mortality.

The predominant risk factor for COPD is tobacco smoking, with other environmental pollutants also known to contribute. Cigarette smoke leads to activation of macrophages and CD8 T lymphocytes that release inflammatory mediators and cytokines. The process also involves neutrophil attraction and cell apoptosis (Barnes 2000). To date, smoking cessation is the only intervention known to slow the decline in lung function associated with COPD (GOLD 2020).

Pharmacotherapy is commonly used to treat people with COPD, with effects on symptoms, quality of life, or frequency and severity of exacerbations (Celli 2004; GOLD 2020). Mainstays of treatment include short‐ and long‐acting inhaled beta₂‐agonists (LABAs) and anticholinergics, corticosteroids, and methylxanthines. Triple therapy with inhaled corticosteroids, LABAs, and long‐acting muscarinic antagonists (LAMAs) can improve FEV₁, reduce exacerbations, and improve patient‐reported outcomes (GOLD 2020). New approaches to treatment are needed, as no individual agent slows the decline in lung function or survival. In the TORCH study (Calverley 2007), a combination of salmeterol 50 μg and fluticasone 500 μg twice daily reduced the risk of death by 17% compared with placebo over the three‐year trial period; however, this finding did not reach the pre‐defined level of statistical significance for the study.

An exacerbation of COPD is an acute and sustained increase in symptoms that results in the need for additional therapy (GOLD 2020). Risk of exacerbation is significantly increased in more severe cases of COPD. Exacerbations have a negative impact on quality of life and lead to more rapid COPD progression, as well as to higher healthcare utilisation and associated costs. A greater impact on health is seen in a subgroup of people with COPD who are more susceptible to exacerbations (defined as "frequent exacerbators"), who have at least two treated exacerbations per year (Le Rouzic 2018).

Common triggers are respiratory viral infection, bacterial infection, and air pollution (Wedzicha 2007; White 2003), which may lead to increased airway inflammation, production of mucus, acute deterioration in lung function, hyperinflation from gas trapping, or a combination of these symptoms (Van Geffen 2015). These processes contribute to symptoms of increased dyspnoea and cough, as well as to changes in the character or volume of sputum.

Description of the intervention

The intervention is an oral medicine that is a selective inhibitor of the isoenzyme phosphodiesterase‐4 (PDE₄). This isoenzyme has a role in airway inflammation and bronchoconstriction, both of which are pathological features of COPD (Boswell‐Smith 2006). Two medicines in this class that have been studied are roflumilast and cilomilast.

How the intervention might work

Cyclic adenosine monophosphate (cAMP) is a secondary messenger that suppresses the activity of inflammatory cells and mediates the process of smooth muscle relaxation in the airways. Phosphodiesterases, in turn, hydrolyse and turn off the biological activity of cAMP (Boswell‐Smith 2006). Therefore, inhibitors of phosphodiesterase action should theoretically provide improvements in the extent of airway narrowing and damage from inflammation.

Non‐selective phosphodiesterase (PDE) inhibitors such as theophylline, a methylxanthine, have been used for years for treatment of people with COPD. These are recommended by current international guidelines as part of adjunctive therapy to long‐acting bronchodilators (GOLD 2020). Limitations to their use include a narrow therapeutic margin and the frequency of adverse effects, which may occur even when the plasma level is within the therapeutic range (Boswell‐Smith 2006). Common adverse effects associated with theophylline include headache, nausea, vomiting, diarrhoea, restlessness, nervousness, insomnia, and gastrointestinal effects (Barnes 2003). Less common, but more serious, are increased risks of cardiac arrhythmia and seizure (Barnes 2003). Some of the adverse effects associated with theophylline have been attributed to its non‐selective PDE inhibition and concurrent adenosine receptor antagonism (Barnes 2005).

The isoenzyme PDE₄ is the predominant isoenzyme involved in metabolising cAMP in immune and inflammatory immune cells, such as neutrophils, macrophages, T cells, and endothelial cells in COPD; and in airway smooth muscle and pulmonary nerves (Agusti 2005; Boswell‐Smith 2006; Torphy 1998; Vignola 2004). Inhibition of PDE₄ leads to elevation of cAMP in inflammatory and immunomodulatory cells, resulting in suppression of inflammatory cell function, relaxation of airways smooth muscle, and modulation of pulmonary nerves (Boswell‐Smith 2006; Essayan 2001; Torphy 1999). Thus, PDE₄ is an attractive target for inhibition in COPD. Furthermore, central nervous system (CNS) and cardiovascular adverse effects experienced by patients treated with the non‐selective PDE inhibitor, theophylline, are the result of adenosine receptor antagonism. This feature is not present with PDE₄‐specific inhibitors (Vignola 2004).

Why it is important to do this review

The development of selective PDE₄ inhibitors offers new hope for therapy offering both anti‐inflammatory and bronchodilatory effects in COPD, with fewer of the adverse effects encountered with non‐selective inhibitors. Additionally, PDE₄ inhibitors may be easier to use because they provide less pharmacokinetic variability and lower potential for drug interactions compared with theophylline (Barnes 2005).

Several PDE₄ inhibitors have been developed, with some progressing to phase 3 clinical trials. These include the second‐generation PDE₄ inhibitors roflumilast (Nycomed, formerly Altana) and cilomilast (GlaxoSmithKline).

Earlier studies of roflumilast have shown significant improvement in pre‐bronchodilator FEV₁ and reduced annual rates of exacerbation among people with severe to very severe COPD who also have chronic bronchitis (Calverley 2009). Roflumilast may be considered in people taking triple inhaled therapy who still have exacerbations, FEV₁ less than 50% predicted, and chronic bronchitis, especially if they have had a hospitalisation in the last year (GOLD 2020).

This review update focuses on effects of PDE₄ inhibitors for treatment of people with stable COPD, using clinically important outcomes. Collating this evidence into a systematic review allows an assessment as to whether or not the theoretical benefits of PDE₄ inhibitors translate into useful clinical effects, and may suggest the potential place of PDE₄ inhibitors within the increasing pharmacopoeia of COPD treatments.

Objectives

To evaluate the efficacy and safety of oral PDE₄ inhibitors for management of stable COPD.

Methods

Criteria for considering studies for this review

Types of studies

We included randomised controlled trials (RCTs) that compared orally administered PDE₄ inhibitors with placebo. We included any long‐term treatment trials but excluded single‐dose trials, as well as trials in acute exacerbations of COPD. We also excluded cross‐over trials to reduce non‐random sources of bias between studies.

Types of participants

Adults (over 18 years of age) with COPD, as defined by the American Thoracic Society, the European Respiratory Society, or GOLD, with airflow obstruction evident by spirometry with post‐bronchodilator FEV₁/FVC of 0.7 or less (GOLD 2020). We considered trials that included participants with both COPD and asthma only if data from participants with COPD could be extracted separately from the study report or through correspondence with the study authors. We excluded ex vivo experiments and trials with participants requiring mechanical ventilation on presentation.

Types of interventions

We included trials if they compared outcomes for participants who received an orally administered PDE₄ inhibitor with those for control participants who received placebo.

Types of outcome measures

Primary outcomes
  • Changes in lung function from baseline including forced expiratory volume in one second (FEV₁), forced vital capacity (FVC), or peak expiratory flow (PEF)

  • Quality of life (e.g. total score on St George's Respiratory Questionnaire (SGRQ))

Secondary outcomes
  • Incidence of COPD exacerbations

  • Symptoms (breathlessness on Borg and other scales and Shortness of Breath Questionnaire; composite measures (summary symptom score))

  • Exercise tolerance (six‐minute walk test)

  • Adverse events (number of participants experiencing one or more adverse event, e.g. gastrointestinal, central nervous system (CNS), and cardiovascular adverse events; change in weight; withdrawal rates)

  • Serious adverse events

  • Mortality

Search methods for identification of studies

Electronic searches

The previously published version included searches up to October 2016. We updated the search for this version from 2016 to 9 March 2020.

We identified trials from the Cochrane Airways Trials Register (Cochrane Airways 2019), which is maintained by the Information Specialist for the Group. The Cochrane Airways Specialised Register contains studies identified from several sources.

  • Monthly searches of the Cochrane Central Register of Controlled Trials (CENTRAL), in the Cochrane Library, through the Cochrane Register of Studies Online (crso.cochrane.org).

  • Weekly searches of MEDLINE Ovid SP 2016 to March 2020.

  • Weekly searches of Embase Ovid SP 2016 to March 2020.

  • Monthly searches of PsycINFO Ovid SP 2016 to March 2020.

  • Monthly searches of the Cumulative Index to Nursing and Allied Health Literature (CINAHL) EBSCO 2016 to March 2020.

  • Monthly searches of the Allied and Complementary Medicine Database (AMED) EBSCO.

  • Handsearches of proceedings of major respiratory conferences.

Studies contained in the Trials Register are identified through search strategies based on the scope of Cochrane Airways. Details of these strategies, as well as a list of handsearched conference proceedings, are provided in Appendix 1. See Appendix 2 for search terms used to identify studies for this review.

We also searched the following trials registries.

  • US National Institutes of Health Ongoing Trials Register ClinicalTrials.gov (www.clinicaltrials.gov/).

  • World Health Organization International Clinical Trials Registry Platform (apps.who.int/trialsearch/).

We searched the Cochrane Airways Trials Register and additional sources to March 2020, with no restriction on language or type of publication. The original strategy for this review, which was more sensitive but less specific, is provided in Appendix 3.

Searching other resources

We checked the reference lists of all primary studies and review articles for additional references and the websites of clinical trials registries for unpublished trial data. We searched relevant manufacturers' websites for study information and PubMed for errata or retractions from included studies published in full text (www.ncbi.nlm.gov/pubmed).

Data collection and analysis

Selection of studies

Two review authors (SJ, RF) independently screened the titles and abstracts of search results and coded them as 'retrieved' (eligible or potentially eligible/unclear) or 'did not retrieve'. We retrieved the full‐text study reports of all potentially eligible studies, and two review authors (SJ, RF) independently screened them for inclusion, recording reasons for exclusion of ineligible studies. We resolved any disagreements through discussion. We identified and excluded duplicates and collated multiple reports of the same study, so that each study, rather than each report, was the unit of interest in the review. We recorded the selection process in sufficient detail to complete a PRISMA flow diagram and Characteristics of excluded studies table (Moher 2009). We categorised references according to trial name (by drug name and number, or by author and year).

Data extraction and management

For the current update, we used an Excel spreadsheet to extract data and assess risk of bias for each included study. One review author (SJ) extracted data on characteristics of included studies (methods, participants, interventions, outcomes) and results of the included studies. We contacted sponsors of the included studies for unpublished data and searched the sponsor's website for further details of outcomes if needed.

We extracted the following data.

  • Methods: study design, total duration of study, details of any 'run‐in' period, number of study centres and locations, study setting, withdrawals, and date of study.

  • Participants: N, mean age, severity of condition, baseline lung function, inclusion criteria, and exclusion criteria.

  • Interventions: intervention, comparison, concomitant medications, and excluded medications.

  • Outcomes: primary and secondary outcomes specified and collected, and time points reported.

  • Notes: funding for studies and notable conflicts of interest of trial authors.

Two review authors (SJ, RF) independently extracted outcome data from the included studies. We noted in the Characteristics of included studies table if outcome data were not reported in a useable way. We resolved any disagreements by consensus. One review author (SJ) transferred data into the Review Manager 5 file (RevMan 2014). We double‐checked that data were entered correctly by comparing data presented in the systematic review against the study reports. A third review author (PP) spot‐checked study characteristics for accuracy against the study report.

Assessment of risk of bias in included studies

Two review authors (SJ, RF) independently assessed risk of bias for each study using the criteria outlined in the Cochrane Handbook for Systematic Reviews of Interventions (Higgins 2019). We resolved disagreements by discussion. We assessed risk of bias according to the following domains.

  • Random sequence generation.

  • Allocation concealment.

  • Blinding of participants and personnel.

  • Incomplete outcome data.

  • Selective outcome reporting.

  • Other bias.

We judged each potential source of bias as high, low, or unclear, and we provided a quote from the study report together with a justification for our judgement in the 'Risk of bias' table. We summarised 'Risk of bias' judgements across different studies for each of the domains listed. We considered blinding separately for different key outcomes when necessary. When information on risk of bias related to unpublished data or correspondence with trialists, we noted this in the 'Risk of bias' table.

When considering treatment effects, we took into account the risk of bias for studies that contributed to that outcome.

Assessment of bias in conducting the systematic review

We conducted the review according to the published protocol and justified any deviations from it in the Differences between protocol and review section of this systematic review.

Measures of treatment effect

The outcomes included in this review were either dichotomous or continuous. For dichotomous outcomes, we recorded the number of participants with one or more outcome events by allocated treatment group.

We undertook meta‐analyses only when this was meaningful, that is, when treatments, participants, and the underlying clinical question were similar enough for pooling to make sense. We expressed results for pooled outcomes with dichotomous variables using a fixed‐effect odds ratio (OR) with 95% confidence interval (CI). Results for continuous variables were expressed as mean differences (MDs) using a fixed‐effect or standardised mean difference (SMD), with 95% CI. We considered a P value less than 0.05 statistically significant. We combined rate ratios on a natural logarithm scale and weighted them by the inverse of the variance of the log rate ratio. We used intention‐to‐treat or 'full analysis set' analyses when they were reported (i.e. analyses for which data had been imputed for participants who were randomly assigned but did not complete the study) instead of completer or per‐protocol analyses.

For change in FEV₁, we used 100 mL as the minimally important difference (MID). For SGRQ, the scale was measured from 0 to 100, with higher scores indicating more limitations. A change in score of 4 units was considered as the MID.

We presented the data as forest plots when possible to show size and direction of effect for treatments with 95% CIs (certainty) using Review Manager 5 (RevMan 2014).

When a single study reported multiple trial arms, we included only the relevant arms. We reported details of the additional arms in the Characteristics of included studies table. When two comparisons (e.g. intervention A versus placebo and intervention B versus placebo) are combined in the same meta‐analysis, we will combine the active arms or will halve the control group to avoid double‐counting.

If adjusted analyses were available (ANOVA or ANCOVA), we used these as a preference in our meta‐analyses. If both change from baseline and endpoint scores were available for continuous data, we used change from baseline unless there was low correlation between measurements among participants. If a study reported outcomes at multiple time points, we used the latest time point. If studies reported post‐treatment follow‐up, we extracted this information and reported it narratively.

Unit of analysis issues

For dichotomous outcomes, we used participants, rather than events, as the unit of analysis (e.g. number of participants experiencing an adverse event rather than the number of adverse events). However, if a study reported rate ratios, we analysed them on this basis.

Dealing with missing data

We contacted the respective pharmaceutical companies for missing trial data. In particular, Nycomed and Forest Laboratories provided us with some study details and results extracted from published articles and abstracts that were not identified in our initial search.

We contacted investigators or study sponsors to verify key study characteristics and to obtain missing numerical outcome data when possible (e.g. when a study was identified as an abstract only). When this was not possible, and missing data were thought to introduce serious bias, we took this into consideration when performing the GRADE assessment for affected outcomes.

Assessment of heterogeneity

We used the I² statistic, along with P values (Higgins 2003), to measure heterogeneity among the trials in each analysis. For I², we employed the following criteria.

  • 0% to 40%: might not be important.

  • 30% to 60%: may represent moderate heterogeneity.

  • 50% to 90%: may represent substantial heterogeneity.

  • 75% to 100%: considerable heterogeneity.

In the case of substantial heterogeneity, we reported it and explored possible causes by conducting pre‐specified subgroup analysis.

Assessment of reporting biases

We compared available published outcomes with prescribed methods and, when available, original study protocols. If we were able to pool more than 10 studies, we created and examined a funnel plot to explore possible small‐study and publication biases.

Data synthesis

We used a fixed‐effect model and performed a sensitivity analysis by using a random‐effects model.

'Summary of findings' tables

We assessed the certainty of evidence for change in FEV₁ lung function, change in quality of life, COPD exacerbations, adverse events, diarrhoea, and all‐cause mortality. We conducted assessments according to recommendations put forth by the GRADE Working Group (Guyatt 2008) and presented in Table 1. We used the five GRADE considerations (risk of bias, consistency of effect, imprecision, indirectness, and publication bias) to assess the quality of a body of evidence related to studies that contributed data for the pre‐specified outcomes. We used the methods and recommendations described in Higgins 2019, employing GRADEpro GDT software (GRADEpro GDT 2015). We justified all decisions to downgrade the certainty of evidence by using footnotes and made comments to aid the reader's understanding of the review when necessary. We applied the clinical importance of results using published minimal important differences (MIDs), when available (e.g. SGRQ has well‐established MIDs in the literature).

Summary of findings 1. Phosphodiesterase‐4 inhibitors compared to placebo for chronic obstructive pulmonary disease.
Phosphodiesterase‐4 inhibitors compared to placebo for chronic obstructive pulmonary disease
Patient or population: people with stable chronic obstructive pulmonary disease
Setting: community‐based, randomised, parallel, double‐blind, placebo‐controlled trials
Intervention: phosphodiesterase 4 inhibitors
Comparison: placebo
Outcomes Anticipated absolute effects* (95% CI) Relative effect
(95% CI) №. of participants
(studies) Certainty of the evidence
(GRADE) Comments
Risk with placebo Risk with PDE₄ inhibitor
Change in FEV₁
Follow‐up: weighted mean 40 weeks Mean FEV₁ was ‐21.37 mL MD 49.33 mL higher
(44.17 higher to 54.49 higher) 20815
(29 RCTs) ⊕⊕⊝⊝
Moderatea,b This is an overall analysis of the outcome that includes roflumilast 250 µg, roflumilast 500 µg, cilomilast 15 mg, and tetomilast 50 µg
MID for FEV₁ is 100 mL
Change in FVC
Follow‐up: weighted mean 45 weeks
Mean FVC was ‐42.47 MD 86.98 higher
(74.65 higher to 99.31 higher)
22108
(17 RCTs)
⊕⊕⊕⊕
High This is an overall analysis of the outcome that includes roflumilast and cilomilast studies
Change in PEF
Follow‐up: weighted mean 42 weeks
Mean PEF was ‐2.82 MD 6.54 higher (3.95 higher to 9.13 higher) 4245
(5 RCTs)
⊕⊕⊝⊝
Lowc This is an overall analysis of the outcome that includes roflumilast and cilomilast studies
Change in SGRQ total score
Follow‐up: weighted mean 33 weeks Mean SGRQ total score was ‐2.21 SGRQ units MD 1.06 SGRQ units lower
(1.68 lower to 0.43 lower) 7645
(12 RCTs) ⊕⊕⊕⊝
Moderatea,b This is an overall analysis of the outcome that includes roflumilast 500 µg, roflumilast 250 µg, and cilomilast 15 mg. Lower scores on the SGRQ represent improved quality of life. The MID for this scale is a change of 4 units. This result does not reach the MID for this scale (0 to 100; higher scores indicate more limitations)
Number of participants with 1 or more exacerbations
Follow‐up: weighted mean 40 weeks 33 per 100 27 per 100
(26 to 29) OR 0.78
(0.73 to 0.84) 20382
(27 RCTs) ⊕⊕⊕⊕
High This is an overall analysis of the outcome that includes roflumilast 500 µg, cilomilast 15 mg, and tetomilast 50 µg
Number of participants experiencing an adverse event
Follow‐up: weighted mean 39 weeks 63 per 100 69 per 100
(68 to 71) OR 1.30
(1.22 to 1.38) 21310
(30 RCTs) ⊕⊕⊝⊝
Lowb,c This is an overall analysis of the outcome that includes roflumilast 500 µg, cilomilast 15 mg, and tetomilast 50 µg, and participants who reported COPD exacerbations as an adverse event
Gastrointestinal adverse effects: diarrhoea
Follow‐up: weighted mean 39 weeks 4 per 100 11 per 100
(10 to 12) OR 3.10
(2.74 to 3.50) 20623
(29 RCTs) ⊕⊕⊕⊕
Highb This is an overall analysis of the outcome that includes roflumilast 500 µg, cilomilast 15 mg, and tetomilast 50 µg. Diarrhoea was the most commonly reported gastrointestinal side effect. See Figure 1. Weight loss was more common and may be a result of diarrhoea
Psychiatric adverse effects (roflumilast 500 µg)
Follow‐up: 6 to 52 weeks
3 per 100 7 per 100 (6 to 8) OR 2.13 (1.79 to 2.54) 11168 (14 studies) ⊕⊕⊕⊝
Moderated Pooled data from FDA website, not individual trial reports
Mortality
Follow‐up: weighted mean 40 weeks 1 per 100 1 per 100
(1 to 2) OR 0.98
(0.77 to 1.24) 19786
(27 RCTs) ⊕⊕⊕⊝
Moderatee This is an overall analysis of the outcome that includes roflumilast 500 µg, cilomilast 15 mg, and tetomilast 50 µg
*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).
CI: confidence interval; COPD: chronic obstructive pulmonary disease; FDA: US Food and Drug Administration; FEV₁: forced expiratory volume in 1 second; FVC: forced vital capacity; MD: mean difference; MID: minimally important difference; OR: odds ratio; PEF: peak expiratory flow; PDE₄: phosphodiesterase‐4 inhibitor; RCT: randomised controlled trial; RR: risk ratio; SGRQ: St George's Respiratory Questionnaire.
GRADE Working Group grades of evidence.High certainty: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate certainty: we are moderately confident in the effect estimate: the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low certainty: our confidence in the effect estimate is limited: the true effect may be substantially different from the estimate of the effect.
Very low certainty: we have very little confidence in the effect estimate: the true effect is likely to be substantially different from the estimate of effect.

aThe outcome was downgraded by 1 point due to moderate heterogeneity across studies (I² = 30% to 60%).

bAlthough some publication bias was found on further investigation through a sensitivity analysis, we did not consider the removal of studies suspected of publication bias to have a large enough impact on the overall effect estimate and CIs. Therefore, we did not downgrade for publication bias.

cThe outcome was downgraded by 2 points due to substantial heterogeneity across studies (I² = 50% to 90%).

dBased on data from the combined COPD safety pool. Individual study data not obtained.

eThe outcome was downgraded by 1 point due to a small number of events, leading to wide confidence intervals.

Subgroup analysis and investigation of heterogeneity

We planned to carry out the following subgroup analyses.

  • Severity of airflow obstruction at baseline (FEV₁ % predicted GOLD grade II 50% to 80%, grade III 30% to 50%, grade IV < 30%) (GOLD 2020).

  • Drug (e.g. roflumilast, cilomilast).

  • Dose (e.g. roflumilast 250 μg or 500 μg).

  • Duration of therapy (≤ 12 weeks; 24 to 26 weeks; 52 weeks; > 52 weeks).

  • Concomitant therapy (inhaled or oral corticosteroids, inhaled long‐acting beta₂‐agonists, or anticholinergics, or both).

We used the formal test for subgroup interactions in Review Manager 5 (RevMan 2014).

Sensitivity analysis

We planned to carry out the following sensitivity analyses, removing the following from the primary outcome analyses.

  • Studies with high risk of bias in one or more domains.

We planned to compare results from a fixed‐effect model by using a random‐effects model.

We did not anticipate the large number of unpublished trials at the protocol stage. Consequently, we undertook a sensitivity analysis of effect sizes for the primary outcomes reported in published and unpublished trials.

Results

Description of studies

Results of the search

See Figure 2 for study flow diagram (Moher 2009).

1.

1

Flow diagram.

From the previous updates (2011 to 2016), 37 studies were included in the review (reference to 2011, 2013, and 2016 reviews). From the current database update search (2016‐2020), 261 abstracts were identified, one of which was a duplicate and was removed. Full texts for 42 relevant references were assessed further for inclusion (Table 2). Of 27 references that were selected for inclusion, 18 references were new additional references to already included studies, one of which was a new additional reference to an ongoing study that had already been identified previously. Four unpublished trials met the inclusion criteria; however, the data for these trials were not available (NCT01595750; NCT00671073; NCT01701934; EUCTR2004‐004442‐40‐GB). Five new trials were identified that met the inclusion criteria and were included in the analyses (Kavitha 2018; Liu 2018; NCT00874497 (EMPHASIS); RO‐2455‐402‐RD (ROBERT); Urban 2018 (ELASTIC)).

1. Number of references for which we sought full text.
Search date: No. of references for which we sought full text
December 2008 53
January 2010 5
August 2010 12
June 2013 20
October 2016 28
April 2020 42

Included studies

Details of the 42 studies included in this review are described in detail in the Characteristics of included studies section.

Of the 42 studies, 27 studies examined roflumilast (COPD safety pool; Kavitha 2018; Liu 2018; RO‐2455‐301‐RD (ACROSS); RO‐2455‐402‐RD (ROBERT); RO‐2455‐404‐RD (REACT); Roflumilast DAL‐MD‐01; Roflumilast FK1 101; Roflumilast FK1 103; Roflumilast FLUI‐2011‐77; Roflumilast IN‐108; Roflumilast JP‐706; Roflumilast M2‐107; Roflumilast M2‐110; Roflumilast M2‐111; Roflumilast M2‐111+M2‐112; Roflumilast M2‐112; Roflumilast M2‐118; Roflumilast M2‐119; Roflumilast M2‐121; Roflumilast M2‐124; Roflumilast M2‐124+M2‐125; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND); Urban 2018 (ELASTIC)), 14 trials studied cilomilast (Cilomilast 039; Cilomilast 042; Cilomilast 076; Cilomilast 091; Cilomilast 103657; Cilomilast 110; Cilomilast 111; Cilomilast 121; Cilomilast 156; Cilomilast 157; Cilomilast 168; Cilomilast 180; Cilomilast 181; Compton 2001), and one trial explored the use of tetomilast (NCT00874497 (EMPHASIS)).

Most of the roflumilast trials were funded by pharmaceutical companies including AstraZeneca and GlaxoSmithKline. Three trials did not report funding information (Kavitha 2018; Liu 2018; Roflumilast FK1 103). One study was funded by Ludwig Boltzmann Institute (Urban 2018 (ELASTIC)). All cilomilast studies were funded by GlaxoSmithKline, and one tetomilast study was funded by Otsuka Pharmaceutical Development & Commercialization, Inc (NCT00874497 (EMPHASIS)).

Almost all studies used inclusion criteria of spirometry and a history of smoking. Only 6 of the 39 studies mandated a history of exacerbation in the previous year (Cilomilast 103657; Cilomilast 121; NCT00874497 (EMPHASIS); Roflumilast M2‐124; Roflumilast M2‐125; Urban 2018 (ELASTIC)).

The mean age of participants in these trials ranged from 60 to 70 years, with the proportion of male participants between 49% and 96%. Mean FEV₁ (% predicted) in trials that reported it ranged from 33% to 51%. Most trials included participants at all stages of COPD; however limitation to those with severe and very severe COPD occurred in RO‐2455‐301‐RD (ACROSS), RO‐2455‐404‐RD (REACT), Roflumilast DAL‐MD‐01, Roflumilast M2‐111, Roflumilast M2‐112, Roflumilast M2‐124, Roflumilast M2‐125, and Roflumilast ROF‐MD‐07(RE2SPOND).

Roflumilast studies

Most of the trials were designed as randomised, double‐blind, placebo‐controlled studies, apart from Urban 2018 (ELASTIC), which was triple‐blinded, and Kavitha 2018, which was assumed to have no blinding. All studies before 2013, apart from Roflumilast JP‐706, were included in combined safety figures for roflumilast that have been made available through publications on the FDA website (https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022522Orig1s000MedR.pdf). Combined safety figures also include participants in two other 24‐week studies (Roflumilast M2‐110; Roflumilast M2‐121), for which results have not been published (roflumilast 500 µg: 5970; roflumilast 250 µg: 1002; placebo: 5682).

All studies compared 500 µg of roflumilast in the intervention group with placebo, with the exception of one study, which was an early‐dose selection study comparing participants who were given roflumilast 250 µg and 500 µg for 24 weeks (Roflumilast M2‐107). The duration of roflumilast treatment in studies ranged from 12 to 52 weeks.

The history of roflumilast studies can be explained in order of publication. The first published PDE₄ inhibitor study for COPD treatment was 52 weeks in duration and allowed concomitant corticosteroid use (Roflumilast M2‐112). Subsequently, results of a replicate study were published (Roflumilast M2‐111). Another two studies were completed that investigated the effects of roflumilast for 52 weeks in participants with severe to very severe COPD with associated chronic bronchitis who were at risk of exacerbations (Roflumilast M2‐124; Roflumilast M2‐125).

Two studies evaluated the add‐on use of roflumilast with long‐acting bronchodilator agents (Roflumilast M2‐127; Roflumilast M2‐128), the first with salmeterol and the second with tiotropium. Both studies ran for 24 weeks. A further two studies ‐ RO‐2455‐404‐RD (REACT) and Roflumilast ROF‐MD‐07(RE2SPOND) ‐ added roflumilast or placebo to a fixed‐dose ICS/LABA combination. Roflumilast M2‐118 was a 12‐week study that focused on airway physiology during rest and exercise in participants with moderate to severe disease. Roflumilast M2‐119 investigated pulmonary function and safety in a group of participants recruited at centres across the Asia‐Pacific regions. Roflumilast DAL‐MD‐01 was mainly aimed at investigating effects on sputum and other biomarkers. Roflumilast FLUI‐2011‐77 explored the airway architecture using imaging techniques.

Three more large RCTs were completed ‐ RO‐2455‐301‐RD (ACROSS), RO‐2455‐404‐RD (REACT), and Roflumilast ROF‐MD‐07(RE2SPOND). RO‐2455‐301‐RD (ACROSS) was carried out across three centres in mainland China, Hong Kong, and Singapore and investigated the effects and safety of roflumilast over 24 weeks. Both RO‐2455‐404‐RD (REACT) and Roflumilast ROF‐MD‐07(RE2SPOND) were 52‐week multi‐centre trials investigating effects on rates of moderate and severe exacerbations.

Four additional trials were completed in 2017 and 2018 ‐ Kavitha 2018, Liu 2018RO‐2455‐402‐RD (ROBERT), RO‐2455‐402‐RD (ROBERT), and Urban 2018 (ELASTIC). RO‐2455‐402‐RD (ROBERT) was a multi‐centre study carried out across Denmark, Germany, Poland, Sweden, and United Kingdom for 16 weeks. The primary aim of this study was to investigate effects on inflammatory markers and changes in lung function. Urban 2018 (ELASTIC) was an Austrian study carried out over 26 weeks, primarily to assess effects of subclinical atherosclerosis and markers of inflammation, but also lung function, exercise, and health impact, in participants with stable COPD. Kavitha 2018, a 12‐week single‐centre study in India investigating effects of roflumilast on change in pulmonary function of participants with moderate to severe disease taking a combined LABA and tiotropium metered‐dose inhaler. Liu 2018 was a 52‐week single‐centre study in China that primarily investigated change in lung function among participants with moderate to severe disease.

Two trials were reported only as conference posters: Roflumilast FK1 101 and Roflumilast FK1 103. The first compared roflumilast 500 µg, roflumilast 250 µg, and placebo for 26 weeks; the second compared roflumilast 500 µg once daily for 24 weeks with roflumilast 500 µg once daily for 12 weeks, then with placebo once daily for the following 12 weeks.

Unpublished results were identified for two other studies: Roflumilast IN‐108 compared the safety and efficacy of roflumilast 250 μg and 500 μg in participants recruited from five centres across India; however, no inclusion criteria were stated, concomitant medications were poorly described, and only 15 participants in the placebo group completed the protocol. Roflumilast JP‐706 was a 24‐week study sponsored by a different collaborator that, in addition to treatment effects, monitored pharmacokinetic levels of roflumilast and its metabolite roflumilast‐N‐oxide.

In the three studies that compared 500 µg or 250 µg with placebo, the placebo group was halved to avoid double counting (Roflumilast FK1 101; Roflumilast IN‐108; Roflumilast M2‐107).

NCT02671942 2016 was identified as an ongoing trial ‐ a Chinese study designed to assess whether altering the standard 500‐µg dose improved tolerability of roflumilast. NCT02451540 (reported as ongoing in the 2016 update) was carried out in Belgium to assess effects of roflumilast on lung function (as measured by functional respiratory imaging) in COPD patients taking LABA/LAMA therapy. This study was reported in the trials registry as being terminated early as no new investigational product could be delivered to the study site.

Further information for three unpublished trials could not be found upon contact with authors (NCT00671073; NCT01595750; NCT01701934).

Cilomilast studies

No new studies were identified for the current update.

Data were derived mainly from phase 3 clinical trials and from one phase 2/3 trial. These included unpublished studies. All used a 15‐mg dose twice daily, except for Compton 2001.

Compton 2001 was a parallel, six‐week, dose‐ranging study comparing placebo with 5 mg, 10 mg, and 15 mg of cilomilast, with FEV₁ as the primary outcome. Pivotal efficacy studies included Cilomilast 039, Cilomilast 042,Cilomilast 091, and Cilomilast 156, all of which were 24 weeks in duration. Cilomilast 121 (phase 2/3, 24 weeks), Cilomilast 157 (52 weeks), and Cilomilast 103657 (24 weeks) followed the pivotal efficacy studies and were smaller in sample size.

Cilomilast 039 and Cilomilast 156 were conducted in North America, and Cilomilast 042 and Cilomilast 091 were conducted in the European Union. Here, primary study outcomes were change in FEV₁, lung function, and SGRQ quality of life score. Cilomilast 076,Cilomilast 110,Cilomilast 111, and Cilomilast 168 were supporting studies, all of which lasted less than 24 weeks, with average trial duration of 10.8 weeks, for which neither FEV₁ lung function nor SGRQ was the primary outcome. Cilomilast 180 (18 weeks) had a primary lung function endpoint ‐ functional residual capacity; Cilomilast 181 (13 weeks) assessed the number of inflammatory cells in a bronchial biopsy.

Tetomilast studies

One new tetomilast study was identified ‐ a phase 2a multi‐centre, randomised, double‐blind, placebo‐controlled study that assessed efficacy and safety in patients with emphysema who had at least one previous exacerbation (NCT00874497 (EMPHASIS)). Study duration was 104 weeks, and the dose of tetomilast was 50 µg. The primary outcome was change in FEV₁ (NCT00874497 (EMPHASIS)).

One unpublished study on oglemilast was identified by the search (NCT00671073); however, no further information could be obtained from trial authors on contact.

Excluded studies

We excluded 15 additional references from the 2020 update at full‐text review, as they did not meet the inclusion criteria. We have provided reasons for exclusion of these 15 studies (see Excluded studies).

Risk of bias in included studies

An overview of risk of bias in individual studies is provided in Figure 3; support for judgements for individual studies is provided under Characteristics of included studies.

2.

2

Methodological quality summary: review authors' judgements about each methodological quality item for each included study.

Allocation

We assessed 12 out of 24 roflumilast studies as having low risk of bias for allocation concealment. Information about allocation concealment for cilomilast studies was limited in publications, but we have considered that this is unlikely to be a source of bias because these studies were sponsored, and standard methods would have been used to minimise the risk of selection bias. We therefore judged the risk of selection bias as low, although allocation concealment is marked as unclear in many of these studies. We considered the only study for tetomilast as having low risk of bias for this domain.

Blinding

All studies included in this review were double‐blind RCTs, with the exception of Kavitha 2018, which failed to report blinding. We regarded overall risk of performance bias and detection bias as low.

Incomplete outcome data

The rate of withdrawal and dropout was reported in 28 of the 39 studies and was generally less than 20% for randomly assigned participants. However, two studies reported higher rates of attrition (NCT00874497 (EMPHASIS); Roflumilast M2‐119). NCT00874497 (EMPHASIS) reported that 54% of participants in both tetomilast and placebo groups did not complete treatments. In addition, five more participants in the tetomilast group than in the placebo group discontinued treatment due to adverse events. Similarly, in Roflumilast M2‐119, more participants in the roflumilast group than in the placebo group discontinued (20% versus 8%). We judged these two studies to be at high risk of bias. We judged the remaining nine studies as having unclear risk of bias due to lack of information about the flow of participants throughout the duration of these studies.

Selective reporting

We identified 27 published and 12 unpublished trials. We performed analyses of differences in treatment effect between published and unpublished treatment groups for primary outcomes and reported this information in the subgroup and sensitivity analyses below.

Other potential sources of bias

We did not consider sponsorship as necessarily increasing the risk of bias when studies were well designed.

For some trials, we noted minor differences in baseline characteristics such as age, gender, FEV₁, and smoking history.

Effects of interventions

See: Table 1

Primary outcomes

Change in FEV₁

We included 32 studies in the main analysis (participants = 20,815). Eighteen studies compared roflumilast 500 µg with placebo (RO‐2455‐301‐RD (ACROSS); RO‐2455‐402‐RD (ROBERT); RO‐2455‐404‐RD (REACT); Roflumilast DAL‐MD‐01; Roflumilast FK1 101; Roflumilast FK1 103; Roflumilast FLUI‐2011‐77; Roflumilast IN‐108; Roflumilast M2‐107; Roflumilast M2‐111; Roflumilast M2‐112; Roflumilast M2‐118; Roflumilast M2‐119; Roflumilast M2‐124; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND), three studies compared roflumilast 250 µg with placebo (Roflumilast FK1 101; Roflumilast IN‐108; Roflumilast M2‐107), 10 studies compared cilomilast 15 mg with placebo (Cilomilast 039; Cilomilast 042; Cilomilast 076; Cilomilast 091; Cilomilast 103657; Cilomilast 110; Cilomilast 121; Cilomilast 156; Cilomilast 157; Compton 2001), and one study compared tetomilast 50 µg with placebo (NCT00874497 (EMPHASIS)).

For RO‐2455‐402‐RD (ROBERT), we calculated standard deviations (SDs) using the RevMan calculator and the number of participants in each treatment group. We did not have change from baseline data for each treatment group; therefore, we used the reported mean difference value between groups (0.063) as the MD for the roflumilast group and an MD of zero for the placebo group.

Urban 2018 (ELASTIC) was not included in the meta‐analysis as the data were skewed and were analysed on a log‐scale as a percentage difference. Similarly, Liu 2018 could not be included in the analysis because reporting of standard errors was unclear, and we received no further correspondence from trial authors on request. Kavitha 2018 reported the outcome separately as endpoint data.

Main analysis

Note that an increase in FEV₁ represents an improvement in lung function.

Based on the 32 trials that reported this outcome, results showed improvement in FEV₁ from baseline among PDE₄ inhibitor‐treated participants compared with controls at a mean of 40.17 weeks' duration (mean difference (MD) 49.33 mL, 95% confidence interval (CI) 44.17 to 54.49; participants = 20,815; studies = 32; I² = 45%; moderate‐certainty evidence) (Analysis 1.1; Figure 4; Table 1). Effects on FEV₁ with 500 µg or 250 µg roflumilast, respectively, were improved (roflumilast 500 µg: MD 55.18 mL, 95% CI 48.65 to 61.71; participants = 14,384; studies = 18; I² = 21%; studies = 17; I² = 26%) (roflumilast 250 µg: MD 56.88 mL, 95% CI 24.38 to 89.38; participants = 1033; studies = 3; I² = 0%). Similar improvement was observed with cilomilast 15 mg (MD 38.15 mL, 95% CI 29.41 to 46.90; participants = 5322; studies = 10; I² = 62%). There was only one small study, with wide uncertainty about effects with tetomilast (MD 82.00 mL, 95% CI ‐50.84 to 214.84; participants = 76) (Analysis 1.1).

1.1. Analysis.

1.1

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 1: FEV₁ (by drug)

3.

3

Forest plot of comparison: 1 PDE₄ inhibitor versus placebo (2020 update), outcome: 1.1 FEV₁ (by drug) [mL].

Moderate and high levels of heterogeneity seen amongst roflumilast 500 µg and cilomilast studies, respectively, can be explained in part by investigation of differences between these two PDE₄ inhibitors (for subgroup analyses, see below).

We investigated publication bias amongst the studies included in the analysis (Figure 1). Four studies were outliers and were investigated further (roflumilast 500 µg: Roflumilast FLUI‐2011‐77; Roflumilast IN‐108; roflumilast 250 µg: Roflumilast IN‐108; cilomilast 15 mg: Compton 2001). These studies were small in population size and contributed very little weight to the overall analysis. In addition, removing these studies from the sensitivity analysis did not have a large impact on the overall effect estimate. We did not downgrade the outcome in our GRADE assessment for this reason (Table 1).

4.

4

Funnel plot of comparison: 1 PDE₄ inhibitor versus placebo (2020 update), outcome: 1.1 FEV₁ (by drug) [mL].

Studies not included in the main analysis

Kavitha 2018 reported FEV₁ at endpoint but did not report the units. It is unclear whether the outcome was reported as litres or millilitres, and trial authors reported much greater improvement compared to authors of another study, which reported improvement of 60 mL (see Kavitha 2018 risk of bias assessment for explanation). Trial authors did not respond when contacted for further information.

Change in FVC from baseline

We included 17 trials in the analysis (Cilomilast 039; Cilomilast 042; Cilomilast 091; Cilomilast 103657; Cilomilast 156; Compton 2001; RO‐2455‐301‐RD (ACROSS); RO‐2455‐402‐RD (ROBERT); RO‐2455‐404‐RD (REACT); Roflumilast M2‐107; Roflumilast M2‐112; Roflumilast M2‐119; Roflumilast M2‐124; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND)).

Treatment with a PDE₄ inhibitor was associated with greater change in FVC from baseline compared to placebo (MD 86.98, 95% CI 74.65 to 99.31; participants = 22,108; studies = 17; I² = 0%; high‐certainty evidence) with no heterogeneity amongst the 17 trials (Analysis 1.2; Table 1).

1.2. Analysis.

1.2

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 2: FVC

Studies not included in the main analysis

Kavitha 2018 reported FVC at endpoint but did not report the units. It is unclear whether the outcome was reported as litres or millilitres; therefore, we did not include this study in the analysis. Trial authors did not respond when contacted for further information.

Change in PEF from baseline

We included six studies in the analysis (Compton 2001; Roflumilast FK1 101 (250 µg); Roflumilast FK1 101 (500 µg); Roflumilast M2‐119; Roflumilast M2‐124; Roflumilast M2‐125). Roflumilast FK1 101 compared one placebo group with roflumilast 250 µg or 500 µg; therefore, the number of participants in the placebo group was halved to avoid double counting, and the study was added to the analysis twice to represent higher and lower doses of roflumilast.

Change in PEF was greater with roflumilast treatment overall than with placebo (MD 6.54 L/min, 95% CI 3.95 to 9.13; participants = 4245; studies = 6; I² = 74; low‐certainty evidence) (Analysis 1.3; Table 1). On further analysis of doses, we noted improvement in PEF with roflumilast 500 µg but not with roflumilast 250 µg when compared with placebo. Upon further investigation, when Compton 2001 was taken out of the analysis, the heterogeneity was zero.

1.3. Analysis.

1.3

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 3: PEF

Change in quality of life
St George's Respiratory Questionnaire (SGRQ)

Note that a decrease in SGRQ total score represents improvement in quality of life.

We included 12 studies in the analysis for this outcome (participants = 7645) (Cilomilast 039; Cilomilast 042; Cilomilast 091; Cilomilast 103657; Cilomilast 121; Cilomilast 156; Cilomilast 157; Compton 2001; Roflumilast DAL‐MD‐01; Roflumilast M2‐107 (250 µg); Roflumilast M2‐107 (500 µg); Roflumilast M2‐112).

Roflumilast M2‐107 reported data for 250 µg and for 500 µg roflumilast compared to one placebo group; therefore, the number of participants in the placebo group was halved to avoid double counting, and the study was included in the analysis twice (Analysis 1.4).

1.4. Analysis.

1.4

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 4: SGRQ total score

We noted a small decrease in total score on the SGRQ from baseline to mean 33 weeks' duration among participants treated with PDE₄ inhibitors compared with those given the control intervention (MD ‐1.06 units, 95% CI ‐1.68 to ‐0.43; participants = 7645; studies = 13; I² = 47%; moderate‐certainty evidence (Analysis 1.4; Figure 5; Table 1). Moderate levels of heterogeneity amongst roflumilast and cilomilast studies can be explained further by subgroup analysis (see below).

5.

5

Forest plot of comparison: 1 PDE₄ inhibitor versus placebo (2020 update), outcome: 1.4 SGRQ total score.

Improvement in symptoms (reported as SGRQ symptom score) was uncertain amongst two studies (Roflumilast M2‐107; Compton 2001) (MD ‐1.53 units, 95% CI ‐4.11 to 1.06; participants = 1048; studies = 2; Analysis 1.5).

1.5. Analysis.

1.5

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 5: SGRQ symptom score

We did not include outcome data for Roflumilast M2‐111, as data were provided in the form of a 'repeated measures analysis', and pooled data did not equal the sum of numbers in each of the individual studies. Liu 2018 was also not included in the analysis due to unclear reporting of standard errors and no response from trial authors.

Secondary outcomes

Incidence of COPD exacerbations

We included 27 trials in the analysis (participants = 20,382) (Cilomilast 039; Cilomilast 042; Cilomilast 076; Cilomilast 091; Cilomilast 111; Cilomilast 121; Cilomilast 156; Cilomilast 157; Cilomilast 168; Cilomilast 180; Liu 2018; NCT00874497 (EMPHASIS); RO‐2455‐301‐RD (ACROSS); RO‐2455‐402‐RD (ROBERT); RO‐2455‐404‐RD (REACT); Roflumilast FK1 101 (500 µg); Roflumilast IN‐108; Roflumilast JP‐706; Roflumilast M2‐107; Roflumilast M2‐111+M2‐112; Roflumilast M2‐119; Roflumilast M2‐124; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND); Urban 2018 (ELASTIC); .

Use of PDE₄ inhibitors was associated with a reduction in the numbers of participants experiencing one or more COPD exacerbations at a mean duration of 40 weeks (odds ratio (OR) 0.78, 95% CI 0.73 to 0.84; high‐certainty evidence; Analysis 1.6; Table 1). This is a relative reduction of more than 20% from a representative risk of 33 per 100 on placebo to 27 per 100 on PDE₄ inhibitors over a weighted mean of 40 weeks (Table 1), and the number needed to treat for an additional beneficial outcome (NNTB) was 20 (95% CI 16 to 27) (Figure 6). There was little heterogeneity among trials (I² = 6%), and a reduction in people experiencing COPD exacerbations was seen with both roflumilast and cilomilast. Tetomilast revealed wide uncertainty about the number of participants experiencing one or more COPD exacerbations; only one study contributed to this result (Analysis 1.6).

1.6. Analysis.

1.6

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 6: Number of participants with 1 or more exacerbations (by drug)

6.

6

In the control group, 33 out of 100 people had an exacerbation of COPD over 40 weeks of treatment, compared to 27 (95% CI 26 to 29) out of 100 people in the active treatment group.

Exacerbation rates

Nine studies reported exacerbation rates and the number of exacerbations experienced on average per participant per year (Cilomilast 157; RO‐2455‐402‐RD (ROBERT); Roflumilast M2‐111; Roflumilast M2‐112; Roflumilast M2‐124; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND)) (Analysis 1.7). We observed a small benefit with treatment, representing a 12% reduction in the exacerbation rate (0.88, 95% CI 0.83 to 0.93).

1.7. Analysis.

1.7

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 7: Exacerbation rate (inverse variance)

Roflumilast FK1 101 reported that the probability of experiencing an exacerbation was reduced by 8% with 250 μg of roflumilast and by 48% with 500 μg, although the absolute value was not reported and it was not stated whether this result was significant.

Four studies reported reduction in severe exacerbation rates per participant per year with PDE₄ inhibitor compared with placebo (Cilomilast 039; RO‐2455‐404‐RD (REACT); Roflumilast M2‐124+M2‐125; Roflumilast ROF‐MD‐07(RE2SPOND)). Cilomilast 15 mg resulted in a 45% reduction in severe exacerbations (P = 0.001) (Cilomilast 039). In studies using roflumilast 500 µg, the reduction in the rate of severe exacerbations ranged from 8.5% to 24.3% across studies (Table 3).

2. Studies reporting severe exacerbation rates per patient per year.
Study ID Percentage reduction (treatment vs placebo) Rate ratio (95% CI) P value
Cilomilast 039 45 0.001
RO‐2455‐404‐RD (REACT) 24.3 0.757 (0.601 to 0.952) 0.0175
Roflumilast M2‐124+M2‐125 17 0.82 (0.63 to 1.06) 0.163
Roflumilast ROF‐MD‐07(RE2SPOND) 8.5 0.95 (0.75 to 1.19) 0.635
Symptoms (breathlessness on Borg or other symptom scales)

We included a total of 14 studies (participants = 10,701) that reported results on the Borg Scale (Analysis 1.8), the Shortness of Breath Questionnaire (Analysis 1.9), the Summary Symptom Scale (Analysis 1.10), or the Breathlessness, Cough, and Sputum Scale (BCSS) (Analysis 1.11) (Borg Scale: Cilomilast 039; Cilomilast 042; Cilomilast 091; Cilomilast 111; Cilomilast 156; Cilomilast 180; Shortness of Breath Questionnaire: Roflumilast M2‐127; Roflumilast M2‐128; Summary Symptom Scale: Cilomilast 039; Cilomilast 042; Cilomilast 091; RO‐2455‐404‐RD (REACT); Roflumilast ROF‐MD‐07(RE2SPOND); BCSS: NCT00874497 (EMPHASIS)).

1.8. Analysis.

1.8

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 8: Borg Scale

1.9. Analysis.

1.9

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 9: Shortness of Breath Questionnaire

1.10. Analysis.

1.10

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 10: Summary symptom score

1.11. Analysis.

1.11

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 11: Breathlessness Cough and Sputum Scale (BCSS) (tetomilast 50 µg)

Overall, the mean difference in change from baseline with PDE₄ inhibitor treatment compared with the control intervention on COPD‐related symptoms at mean duration of 21 weeks was small, regardless of the scale used to measure it. The only effect was seen in one trial of cilomilast ‐ for breathlessness scored on a Borg Scale (MD ‐0.19, 95% CI ‐0.33 to ‐0.05) (Analysis 1.8). This is a small absolute difference so is of doubtful clinical relevance. Results showed no difference with PDE₄ inhibitor in effects on the Summary Symptom Scale (standardised mean difference (SMD) ‐0.02, 95% CI ‐0.07 to 0.03; participants = 6186; studies = 5; I² = 19%), the Shortness of Breath Questionnaire (MD ‐1.09, 95% CI ‐2.47 to 0.28; participants = 1633; studies = 2; I² = 81%), or the BCSS (Analysis 1.11).

Exercise tolerance (six‐minute walk test)

We included six studies that reported the six‐minute walk test (6MWT) (participants = 2055) (Cilomilast 039; Cilomilast 042; Cilomilast 091; Cilomilast 111; Roflumilast DAL‐MD‐01; Urban 2018 (ELASTIC)).

Exercise tolerance was measured on the 6MWT in six trials (two roflumilast and four cilomilast trials). We found uncertainty in walk test distance at a mean duration of 21 weeks between PDE₄ inhibitor and placebo groups (MD 3.50; 95% CI ‐5.84 to 12.85) (Analysis 1.12).

1.12. Analysis.

1.12

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 12: 6‐minute walk test

Adverse events

We included 30 studies in the overall analysis (participants = 21,310) (Cilomilast 039; Cilomilast 042; Cilomilast 076; Cilomilast 091; Cilomilast 103657; Cilomilast 110; Cilomilast 111; Cilomilast 121; Cilomilast 156; Cilomilast 157; Cilomilast 168; Cilomilast 180; Cilomilast 181; Compton 2001; NCT00874497 (EMPHASIS); RO‐2455‐301‐RD (ACROSS); RO‐2455‐402‐RD (ROBERT); RO‐2455‐404‐RD (REACT); Roflumilast DAL‐MD‐01; Roflumilast FK1 101; Roflumilast IN‐108; Roflumilast JP‐706; Roflumilast M2‐107; Roflumilast M2‐111+M2‐112; Roflumilast M2‐119; Roflumilast M2‐124+M2‐125; Roflumilast M2‐127; Roflumilast M2‐128; Roflumilast ROF‐MD‐07(RE2SPOND); Urban 2018 (ELASTIC)).

Overall, the likelihood of a participant experiencing an adverse event at a mean duration of 38 weeks was higher with PDE₄ inhibitor treatment than with placebo (OR 1.30, 95% CI 1.22 to 1.38; participants = 21,310; studies = 30; I² = 64%; low certainty evidence) (Analysis 1.13; Table 1). This effect was seen for both roflumilast and cilomilast but not for tetomilast, as we found only one study for this PDE₄ inhibitor.

1.13. Analysis.

1.13

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 13: Number of participants experiencing an adverse event

Adverse events: roflumilast 500 µg versus roflumilast 250 µg

The higher dose of roflumilast (500 μg) was associated with more adverse events than the lower dose (250 μg); however, this finding was based on only four trials and confidence intervals were wide (OR 1.21, 95% CI 1.01 to 1.46) (Analysis 1.14).

1.14. Analysis.

1.14

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 14: Number of participants experiencing an adverse event (roflumilast 500 μg vs 250 μg)

We found a range of adverse effects that occurred more frequently in PDE₄ inhibitor‐treated participants, which are described below.

Gastrointestinal adverse effects (diarrhoea, nausea, vomiting, dyspepsia, weight loss)

Diarrhoea was more commonly experienced in PDE₄ inhibitor‐treated groups than in placebo groups (OR 3.10, 95% CI 2.74 to 3.50; participants = 20,623; studies = 29; I² = 12%; high‐certainty evidence) (Analysis 1.15; Table 1).

1.15. Analysis.

1.15

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 15: Diarrhoea

Nausea was also reported as an increased side effect (OR 3.79, 95% CI 3.24 to 4.43; participants = 20,949; studies = 28; I² = 24%) (Analysis 1.16), as were vomiting (OR 3.95, 95% CI 2.78 to 5.60; participants = 5986; studies = 12; I² = 0%) (Analysis 1.17) and dyspepsia (OR 3.17, 95% CI 2.33 to 4.30; participants = 6247; studies = 13; I² = 0%) (Analysis 1.18). Weight loss was commonly reported and was an increased adverse effect (OR 3.72, 95% CI 3.09 to 4.47; participants = 12,462; studies = 12; I² = 24%) (Analysis 1.19).

1.16. Analysis.

1.16

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 16: Nausea

1.17. Analysis.

1.17

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 17: Vomiting

1.18. Analysis.

1.18

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 18: Dyspepsia

1.19. Analysis.

1.19

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 19: Weight loss

More than 10% of participants in the PDE₄ inhibitor group experienced gastrointestinal side effects; diarrhoea was the most frequently reported symptom (Figure 7) (number needed to treat for an additional harmful outcome (NNTH) 14, 95% CI 12 to 17).

7.

7

In the control group, 4 out of 100 people had a diarrhoea episode over 39 weeks of treatment, compared to 11 (95% CI 10 to 12) out of 100 people in the active treatment group.

Withdrawals

An increase in withdrawals attributed to adverse events was recorded for both roflumilast and cilomilast treatment groups (OR 1.89, 95% CI 1.73 to 2.07; participants = 21,358; studies = 31; I² = 21%) (Analysis 1.20).

1.20. Analysis.

1.20

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 20: Withdrawals due to adverse events

Headache

We found 23 studies that reported a higher proportion of participants experiencing headache as an adverse effect when taking a PDE₄ inhibitor (OR 1.69, 95% CI 1.46 to 1.94; participants = 19,215; I² = 23%) (Analysis 1.21). Participants in the roflumilast 500 µg treatment group were more likely to experience headache than those given placebo (OR 2.13, 95% CI 1.74 to 2.59; participants = 13,565; studies = 12; I² = 0%) (Analysis 1.21).

1.21. Analysis.

1.21

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 21: Headache

Abdominal pain

Fifteen studies reported abdominal pain as an adverse effect in the PDE₄ inhibitor treatment group (OR 2.02, 95% CI 1.62 to 2.52; participants = 8329; studies = 15; I² = 0%) (Analysis 1.22). A greater likelihood of abdominal pain was noted for participants in the roflumilast treatment group compared with the placebo group (OR 2.77, 95% CI 1.38 to 5.56; participants = 2641; studies = 3). Participants were also more likely to experience abdominal pain with cilomilast than with placebo (OR 1.97, 95% CI 1.55 to 2.49; participants = 5604; studies = 11), although the magnitude of effect was smaller compared to that seen with roflumilast (Analysis 1.22).

1.22. Analysis.

1.22

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 22: Abdominal pain

Influenza‐like symptoms

There was uncertainty in the incidence of influenza‐like symptoms between PDE₄ inhibitors (OR 1.09, 95% CI 0.87 to 1.36; participants = 11,460; studies = 10), as confidence intervals crossed the line of no effect (Analysis 1.23).

1.23. Analysis.

1.23

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 23: Influenza‐like symptoms

Upper respiratory tract infection

There was uncertainty in the incidence of upper respiratory tract infection between PDE₄ inhibitor and placebo treatment groups (OR 0.91, 95% CI 0.81 to 1.04; participants = 17,022; studies = 23), as confidence intervals crossed the line of no effect (Analysis 1.24).

1.24. Analysis.

1.24

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 24: Upper respiratory tract infection

Psychiatric adverse effects: COPD safety pool

We recorded the number of psychiatric adverse events from pooled data derived from all parallel‐design, double‐blind studies investigating roflumilast collated and presented to the FDA. This included data from the 15 fully published trials but excluded results from Roflumilast JP‐706, which was conducted by a different study collaborator. These results reported symptoms of depression separately from depressed mood, depressive symptoms, or major depression. The likelihood of experiencing a psychiatric adverse event was greater in the roflumilast 500 μg treatment group than in the placebo group (OR 2.13, 95% CI 1.79 to 2.54) (Analysis 1.25; Table 1). This was reported in three out of 100 people in the placebo group compared to seven out of 100 in the PDE₄ inhibitor‐treated group (95% CI 6 to 8) (NNTH 28, 95% CI 21 to 39). The likelihood of experiencing a psychiatric adverse event was uncertain with 250 µg roflumilast compared with placebo, as the confidence interval crossed the line of no effect (OR 0.87, 95% CI 0.56 to 1.33) (Analysis 1.25).

1.25. Analysis.

1.25

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 25: Psychiatric adverse events (roflumilast)

An increase in symptoms of anxiety (OR 1.81, 95% CI 1.26 to 2.62) (Analysis 1.26) and depression (OR 1.59, 95% CI 1.11 to 2.27) (Analysis 1.27) was associated with roflumilast 500 µg compared with placebo. Uncertainty about symptoms of anxiety (OR 0.94, 95% CI 0.40 to 2.21) or depression (OR 0.56, 95% CI 0.20 to 1.56) was greater with roflumilast 250 µg compared with placebo, as confidence intervals crossed the line of no effect in both analyses (Analysis 1.26; Analysis 1.27).

1.26. Analysis.

1.26

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 26: Anxiety or anxiety disorder (roflumilast)

1.27. Analysis.

1.27

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 27: Depression (roflumilast)

Three reports described completed suicides and two suicide attempts in roflumilast‐treated participants compared to none in participants given placebo (roflumilast COPD safety database, n = 12,054).

In more recent roflumilast trials, the numbers of participants experiencing insomnia and sleep disorders taking roflumilast 500 µg were greater than among those taking placebo (OR 2.67, 95% CI 2.11 to 3.38) (Analysis 1.28), but results with 250 µg roflumilast were uncertain, as the confidence interval crossed the line of no effect (Analysis 1.28).

1.28. Analysis.

1.28

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 28: Insomnia and sleep disorders (roflumilast)

Serious adverse events

Treatment was found to have no effect on serious adverse events (OR 0.99, 95% CI 0.91 to 1.07; participants = 19,191; studies = 29; I² = 54%) (Analysis 1.29).

1.29. Analysis.

1.29

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 29: Serious adverse events

Mortality

Mortality was a relatively rare event during these trials, results showed no effect of treatment for this outcome (OR 0.98, 95% CI 0.77 to 1.24; participants = 19,786; studies = 27; I² = 0%; moderate‐certainty evidence) (Analysis 1.30; Table 1).

1.30. Analysis.

1.30

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 30: Mortality

Subgroup and sensitivity analyses

Primary outcome: FEV₁

A moderate but significant level of heterogeneity was evidence for the change in FEV₁ outcome when all trials were pooled (I² = 45%). We analysed the data further by performing subgroup and sensitivity analyses.

Subgroup analysis: COPD severity

To see whether the size of the treatment effect varied with COPD severity, we conducted subgroup analyses of trials for which the mean per cent predicted FEV₁ at baseline was available (Analysis 1.31). Effects seen in both old GOLD grade I or II (FEV₁ ≥ 50%) predicted and old GOLD grade III or IV (FEV₁ < 50%) were statistically significant and of similar magnitude (MD 52.78, 95% CI 46.73 to 58.83; test for subgroup differences: Chi² = 0.03, df = 1 (P = 0.87)).

1.31. Analysis.

1.31

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 31: FEV₁ (by mean COPD severity)

Subgroup analysis: dose (roflumilast 500 µg versus roflumilast 250 µg)

For dose effects of roflumilast, both roflumilast 500 µg and roflumilast 250 µg were associated with a similar change in FEV₁ (roflumilast 500 µg: MD 55.18, 95% CI 48.65 to 61.71; participants = 14,384; studies = 18; I² = 21%; roflumilast 250 µg: MD 56.88, 95% CI 24.38 to 89.38; participants = 1033; studies = 3; I² = 0%) (test for subgroup differences: Chi² = 0.01, df = 2 (P = 0.92)) (Analysis 1.1; Figure 4).

Subgroup analysis: duration of treatment

For FEV₁, the size of the treatment effect, that is, the mean difference between PDE₄ and placebo groups, was numerically greater in short studies of 6 to 12 weeks (MD 101.71, 95% CI 70.96 to 132.46; participants = 1191; studies = 8) than in studies of 24 to 26 weeks (MD 46.14, 95% CI 38.44 to 53.84; participants = 8086; studies = 13) and studies of 52 weeks (MD 48.77, 95% CI 41.44 to 56.10; participants = 10,662; studies = 7). However this difference between subgroups may be a chance finding (test for subgroup differences: Chi² = 5.11, df = 6 (P = 0.53)) (Analysis 1.33).

1.33. Analysis.

1.33

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 33: FEV₁ (by study duration)

Subgroup analysis: concomitant therapies (roflumilast versus cilomilast)

With respect to PDE₄ inhibitor use with concomitant therapies (Analysis 1.34), the largest increases in FEV₁ were seen in two trials where participants were taking regular, long‐acting bronchodilators: in one trial, salmeterol (Roflumilast M2‐127), and in the other, tiotropium (Roflumilast M2‐128) (MD 60.52 mL, 95% CI 40.57 to 80.46). The next largest improvements were seen in trials for which all concomitant medications (including long‐acting bronchodilators if previously received) were continued (RO‐2455‐301‐RD (ACROSS); RO‐2455‐404‐RD (REACT); Roflumilast ROF‐MD‐07(RE2SPOND) (MD 56.58 mL, 95% CI 46.91 to 66.25) (Analysis 1.34). A similar improvement in FEV₁ was seen when participants were taking corticosteroids (MD 42.26 mL, 95% CI 25.46 to 59.05) (Analysis 1.34). Improvements in FEV₁ were also noted in trials where only a PDE₄ inhibitor was taken (apart from short‐acting beta₂ agonists) (MD 44.80 mL, 95% CI 37.69 to 51.91) (test for subgroup differences: Chi² = 5.61, df = 3 (P = 0.13) (Analysis 1.34).

1.34. Analysis.

1.34

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 34: FEV₁ (additional medication)

Sensitivity analysis

Using a random‐effects model made no difference in levels of statistical significance nor degree of heterogeneity for the change in FEV₁ (MD 51.49, 95% CI 42.87 to 60.10; Analysis 1.35). Too many 'Risk of bias' domains were judged to be at 'unclear' risk of bias for subgroup analysis to be conducted according to study quality. Of note, some effect sizes were greater in the published trials, for example, the treatment effect on FEV₁ was MD 55.75 mL (95% CI 49.45 to 62.06) in the 20 published trials, and MD 35.05 (95% CI 25.70 to 44.40) in the nine unpublished trials (Analysis 1.36), which was significantly different (test for subgroup differences: Chi² = 12.94, df = 1 (P = 0.0003)). This is illustrated in the funnel plot, with more unpublished studies showing a smaller treatment effect (Figure 8).

1.35. Analysis.

1.35

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 35: FEV₁ (random‐effects model)

1.36. Analysis.

1.36

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 36: FEV₁ (published vs unpublished)

8.

8

Funnel plot of comparison: 2 PDE₄ inhibitor versus placebo (2020 update), outcome: 2.36 FEV₁ (published versus unpublished).

By visual analysis of the forest plot and sequential elimination, we identified the six‐week Compton 2001 cilomilast trial as a major contributor to the heterogeneity of pooled FEV₁ results. When this trial was removed, the overall I² statistic decreased from 45% to 26%, and in the cilomilast subgroup from 62% to 0%. It is notable that this study had the shortest treatment duration (six weeks) and showed the greatest improvement from baseline in FEV₁ lung function in the treatment group across all studies.

Primary outcome: SGRQ
Subgroup analysis: COPD severity

Although quality of life was improved in participants with GOLD grade I or II COPD severity, and with GOLD grade III or IV COPD severity (MD ‐1.56 units, 95% CI ‐2.39 to ‐0.74; participants = 4851; studies = 8) (test for subgroup differences: Chi² = 0.02, df = 1 (P = 0.89)) (Analysis 1.37), overall heterogeneity was high (I² = 55%). Studies in which participants had grade I or II COPD severity were similar (I² = 0%), but variation was observed amongst studies in which participants had grade III or IV COPD severity (I² = 73%) (Analysis 1.37).

1.37. Analysis.

1.37

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 37: SGRQ total score (by mean COPD severity)

Subgroup analysis: duration of treatment

It is notable that in two trials with a duration of one year that reported total SGRQ, the change in quality of life seen with treatment compared with control was uncertain (MD 0.26, 95% CI ‐1.18 to 1.69) (Analysis 1.38). However, quality of life was improved among participants taking a PDE₄ inhibitor for less than 12 weeks (MD ‐4.19, 95% CI ‐7.60 to ‐0.78) and for 24 to 26 weeks (MD ‐1.18, 95% CI ‐1.94 to ‐0.42). A significantly greater treatment effect was noted in short studies (6 to 12 weeks) compared with studies of 24 to 52 weeks. A high level of heterogeneity (I² = 57%) was observed amongst studies providing 24 to 26 weeks of treatment (test for subgroup differences: Chi² = 6.50, df = 2 (P = 0.04)) (Analysis 1.38).

1.38. Analysis.

1.38

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 38: SGRQ total score (by duration)

Sensitivity analysis

Analysis revealed a difference in effect size of the total SGRQ score between published and unpublished trials (MD ‐1.98, 95% CI ‐3.07 to ‐0.89 versus MD ‐0.43, 95% CI ‐1.26 to 0.40) (test for subgroup differences: Chi² = 4.94, df = 1 (P = 0.03)) (Analysis 1.39).

1.39. Analysis.

1.39

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 39: SGRQ total score (by published vs unpublished)

Secondary outcome: exacerbations
Subgroup analysis: additional medications

When investigating whether other additional medication made any difference, we found similar efficacy for both roflumilast and cilomilast, specifically when use of concomitant long‐acting bronchodilators was permitted (OR 0.79, 95% CI 0.73 to 0.85) (test for subgroup differences: Chi² = 1.53, df = 3 (P = 0.67)) (Analysis 1.40).

1.40. Analysis.

1.40

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 40: Number of participants on roflumilast with 1 or more exacerbations (additional medication)

Discussion

Summary of main results

This systematic review evaluated randomised controlled trials (RCTs) that assessed the efficacy and safety of oral phosphodiesterase 4 (PDE₄) inhibitors in people with chronic obstructive pulmonary disease (COPD). The conclusions of this review remain unchanged following the addition of new studies for the 2020 update, reporting small improvements in lung function and quality of life and decreased exacerbations.

Lung function

Based on data from 32 trials (low‐certainty evidence), we found that both roflumilast and cilomilast led to greater improvements in lung function from baseline, as measured by forced expiratory volume in one second (FEV₁), forced vital capacity (FVC), or peak expiratory flow rate (PEF), compared with placebo (Table 1). Furthermore, improvement in lung function was seen regardless of the severity of the disease. This improvement in FEV₁ lung function occurred whether or not PDE4 inhibitor treatment was given in addition to other COPD treatments, such as long‐acting beta₂‐agonists (LABAs) or anticholinergics or inhaled corticosteroids (ICSs).

Greater improvement in FEV₁ was observed in studies of shorter duration (Analysis 1.33); this could be due to a limited short‐term response to PDE₄ inhibitor treatment, as might be seen with tachyphylaxis, and needs further investigation.

The mean change in FEV₁ was less than what is usually considered a minimum clinically important difference (MCID) (100 mL; Donohue 2005), but it was comparable to that seen with other COPD treatments in recent large RCTs. For example, mean improvement in FEV₁ of 49 mL with treatment, as seen in moderate to severe COPD in this review, is of similar magnitude to that seen with fluticasone (47 mL), salmeterol (42 mL), and fluticasone and salmeterol combined (92 mL) in the TORCH 2007 study among people with severe COPD.

Quality of life

Data show only a small improvement in quality of life as assessed by St George's Respiratory Questionnaire (SGRQ) total score. Quality of life had been chosen as a primary outcome because of concerns as to whether or not the adverse effects of PDE₄ inhibitors might outweigh any beneficial COPD‐related events. The average change in SGRQ total score was 1.06 units (over a duration between 6 and 12 months) (Table 1) and was of similar magnitude among trials of participants with milder or more severe COPD. Although this improvement was statistically significant, a change of greater than four units is usually regarded as the MCID (Jones 2005). Although symptom scores were marginally better in the treatment groups, no change was seen in exercise tolerance, suggesting that improvements in respiratory symptoms may not necessarily translate into enhanced physical functioning. Fewer trials were assessable for these outcomes, raising the possibility of type 1 or type 2 error.

Exacerbations

A second major finding, based on data from 27 trials (moderate‐certainty evidence; Table 1), was that participants were more likely to be exacerbation‐free while being treated with PDE₄ inhibitors compared with those given control interventions. Overall, participants were 22% less likely to have an exacerbation, translating to a number needed to treat for an additional beneficial outcome (NNTB) of around 20 (95% confidence interval (CI) 16 to 26) for one person to be exacerbation‐free in the study period (Figure 6; Table 1). Although the likelihood of an individual experiencing an exacerbation was lowered with PDE₄ inhibitor treatment, the decrease in the overall rate of exacerbations was less marked, with a relative reduction of 13%.

Taken together, results for lung function and exacerbations suggest that PDE₄ inhibitors in people with COPD are acting independently of other treatments, particularly bronchodilators. This is an encouraging finding that could be consistent with a broad anti‐inflammatory effect (Fabbri 2009). On the other hand, short‐duration studies showed more favourable results than longer studies, but the reasons for this are unclear. Significant heterogeneity was noted among trials, suggesting that unmeasured differences between trials may be having an impact.

Adverse events

Adverse events were more likely among roflumilast‐ and cilomilast‐treated participants than among those receiving placebo (very low‐certainty evidence; Table 1), particularly gastrointestinal effects such as diarrhoea, nausea, vomiting, and dyspepsia.

Participants in treatment groups were more likely to withdraw from trials because of adverse events; on average, 14% in the treatment groups withdrew compared with 8% in the control groups. Similarly, there was a slight excess in the total numbers of participants in the treatment groups experiencing any adverse event compared with numbers in the control groups (Analysis 1.13). As this analysis included symptoms as well as exacerbations, which were reduced among treatment groups, the analysis will tend to underestimate the excess of non‐COPD‐related adverse events occurring with PDE₄ inhibitor treatment.

It is notable that treatment with roflumilast was associated with an increased incidence of weight loss. Whether this was due to anorexia from gastrointestinal adverse effects or from another effect is not yet clear. Also not clear is whether cilomilast has the same effect, as this has not been studied. Weight loss may be a beneficial effect for people with COPD who are obese. In contrast, low body mass in the later stages of COPD is associated with a worse prognosis and is notoriously difficult to reverse (GOLD 2020). This adverse effect warrants further investigation. It is reassuring that there was no increase in serious adverse events nor in mortality, although trials were of relatively short duration and analyses were underpowered to report on the latter outcome.

Although the lower dose (250 μg) of roflumilast produced similar improvements in FEV₁ (Analysis 1.32) and was associated with slightly fewer adverse events than the larger dose (Analysis 1.14), the lower dose was associated with a smaller reduction in rates of exacerbation when compared with the higher dose in the only trial that reported this (Roflumilast FK1 101). Moreover, data on the lower dose were available from a limited number of studies, and this has not been studied as add‐on therapy to other bronchodilators.

1.32. Analysis.

1.32

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 32: FEV₁ (roflumilast 500 μg vs 250 μg)

Awareness of the risk of psychiatric adverse events associated with roflumilast treatment is growing (Analysis 1.25; Table 1), in particular the increased likelihood of experiencing sleep disturbances, anxiety, and depressed mood. It should be noted that we found three reports of completed suicides and two of suicide attempts among roflumilast‐treated participants compared to none in participants given placebo (roflumilast COPD safety database).

Mortality

Mortality was a rare event, and there was no difference between participants treated with a PDE₄ inhibitor and those given placebo (Analysis 1.30; Table 1).

Overall completeness and applicability of evidence

We have reviewed all known published and unpublished trials identified through standard Cochrane searches, as well as those obtained from the trials register for the National Institutes of Health (NIH) and from pharmaceutical websites.

We have not been able to verify the pooled endpoint data for psychiatric (treatment possibly harmful) and cardiovascular adverse events (treatment possibly beneficial), as we obtained this information from reports on the US Food and Drug Administration (FDA) website and from White 2013, respectively.

To ensure that our Cochrane systematic review accurately reflects all known outcomes of roflumilast therapy, for previous updates we approached the manufacturer of roflumilast for study‐level data on each of the cardiovascular outcomes (cardiovascular death, non‐fatal myocardial infarction, and non‐fatal stroke), as well as the composite outcome, major adverse cardiovascular events (MACE). This would have allowed us to perform comparisons both within (i.e. between roflumilast and placebo groups) and among the studies. Unfortunately, our request for individual trial data was refused, with the following reasons cited.

  • It is inappropriate from a statistical perspective to look into individual trials with too small a sample size for this kind of relatively rare endpoint.

  • It was part of the retrospective analyses to evaluate the whole data set with a sufficiently broad database and not to go into per‐study data that would comprise numbers in each treatment arm that were too low for conclusive interpretation.

  • In none of the studies was blinded adjudication of events implemented as a prospective analysis, which would have required a data release in terms of transparency for each individual study (this is why it was not mentioned in the original publications of individual trials).

In response to the statement by representatives of Takeda Pharmaceuticals Limited, we have urged that these issues be reconsidered for future studies, and that study data be made more widely available. Finally, caution must be used when interpreting associations between COPD exacerbations and MACE, because although treatment groups were matched at baseline, it cannot be assumed that these groups are equivalent when the focus is only on groups of participants who experienced exacerbations. These concerns could not be assessed in this review, as further study data were not provided.

Certainty of the evidence

For the key outcomes of changes in lung function and quality of life, greater beneficial effects of PDE₄ inhibitors were reported in published than in unpublished studies, raising concerns about publication bias. When investigating publication bias further for each outcome, we found that eliminating from the analyses studies with suspected publication bias did not significantly alter overall effect estimates or confidence intervals for lung function or quality of life. Similarly, this was apparent when adverse events were investigated.

We identified a moderate level of heterogeneity for both of the primary outcomes for this review, which is not fully explained by subgroup or sensitivity analyses according to study duration or concomitant medication use. This suggests that unknown factors that may impact effect size have led us to downgrade the quality of evidence and the certainty of our findings (Table 1). In contrast, the blinded design of studies comparing roflumilast or cilomilast with placebo protected against detection bias in our view. The certainty of evidence for a reduction in exacerbation was therefore higher for this comparison. On balance, we believe the true beneficial effect of PDE₄ inhibitors is likely to be no greater than we have reported and is probably less; equally, the harms of PDE₄ treatment may have been understated (due in part to higher withdrawal rates in active treatment arms). On the other hand, as subgroup analyses for COPD severity are based on the mean predicted lung function for the study group and not for individual participants, we cannot rule out benefit for individuals of a specific COPD phenotype.

Addition of new trials

The 2020 update of this review included four studies on roflumilast 500 µg and one study on tetomilast (Kavitha 2018; Liu 2018; NCT00874497 (EMPHASIS); RO‐2455‐402‐RD (ROBERT); Urban 2018 (ELASTIC)). Data from these new trials did not affect the results already yielded by analyses. Kavitha 2018 and Liu 2018 were not included in the analyses for lung function, as the units for this endpoint were unclear even though we contacted trial authors for clarification. Data from Liu 2018 were not included in the meta‐analysis for SGRQ due to unclear data units in the publication.

Potential biases in the review process

Potential biases in the review process were minimised by double‐checking of data extraction and input. The review authors have no conflicts of interest to declare.

Agreements and disagreements with other studies or reviews

Several other meta analyses have been conducted, including Luo 2016, Yuan 2016, and Shen 2018. Each of these included fewer studies than the present review but presented findings and conclusions that were similar. Our findings are also similar to those presented by Wedzicha 2016 and show effects on exacerbations similar to those described in Rabe 2017.

In a post hoc pooled analysis (n = 5595) of four trials in this review (Rennard 2014), roflumilast was seen to improve transition dyspnoea index (TDI) focal scores of breathlessness versus placebo at week 52 (treatment difference 0.327; P < 0.0001). Roflumilast was associated with more TDI responders and fewer TDI deteriorators (≥ 1‐unit increase or decrease from baseline, respectively) versus placebo at week 52 (P < 0.01, both). Rates of MACE in COPD participants treated with PDE₄ inhibitors have been meta‐analysed and reported in White 2013. This review found that risk of cardiovascular death, non‐fatal myocardial infarction, or non‐fatal stroke, when combined into a composite outcome MACE, was reduced in the roflumilast group compared with the placebo group (hazard ratio 0.65, 95% CI 0.45 to 0.93; P = 0.019). On the other hand, hazard ratios for treatment effects for each of these types of events individually were statistically different. Cardiovascular events were higher among participants with baseline cardiovascular risk factors than among those without baseline cardiovascular risk (defined as the presence of hypertension, diabetes mellitus, hyperlipidaemia, and/or a history of heart disease). In addition, it was found that the difference between treatment and placebo was statistically significant only for the group of participants without baseline risk factors. Event rates in a subgroup of trials that were one year in duration show no significant differences between treatment and placebo groups in the proportion of participants who reported a MACE, even when divided into those who did or did not experience a COPD exacerbation. Similarly, between participants with and without MACE events, the proportions of participants experiencing exacerbations were similar (43.2% and 42.1%, respectively).

Authors' conclusions

Implications for practice.

Phosphodiesterase‐4 (PDE₄) inhibitors are oral medicines that may be taken in combination with other standard chronic obstructive pulmonary disease (COPD) treatments. Most evidence has been gathered for roflumilast at a dose of 500 μg daily and cilomilast at 15 mg twice daily.

PDE₄ inhibitors join an increasing list of treatments for COPD that improve short‐term lung function and reduce exacerbations, but they have not been shown to increase life expectancy. Most trials to date have been one year in duration (with the exception of one study of nearly two years' duration). In contrast to long‐acting bronchodilators, PDE₄ inhibitors have minimal benefit for symptoms on a day‐to‐day basis, or for quality of life, and are often associated with adverse effects, especially gastrointestinal effects and headaches. Roflumilast is associated with greater weight loss and increased psychiatric symptoms compared with placebo. Findings of this review provide cautious support for the use of PDE₄ inhibitors in COPD. In accordance with GOLD 2020 guidelines, PDE₄ inhibitors may have a place as add‐on therapy for a subgroup of people with persistent symptoms or exacerbations despite optimal COPD management (e.g. people who are not controlled on fixed‐dose long‐acting beta₂‐agonist (LABA) and inhaled corticosteroid (ICS) combinations).

Implications for research.

This review has highlighted several possible topics for further study.

  • Effects of PDE₄ inhibitors on forced expiratory volume in one second (FEV₁) decline and mortality in studies of longer duration.

  • Effects of PDE₄ inhibitors at shorter time points in longer‐duration studies on FEV₁.

  • Subgroup analysis of participants with/without chronic bronchitis and with/without a history of exacerbations.

  • Effects of PDE₄ inhibitors among participants with frequent exacerbations.

  • Effects of PDE₄ inhibitors on healthcare utilisation, including hospitalisation (incidence and bed days).

  • Direct comparison of PDE₄ inhibitors and inhaled corticosteroids (ICSs) when used as add‐on therapies to tiotropium, to long‐acting beta₂‐agonists (LABA), or to all three (triple inhaled therapies).

  • Direct comparison of tiotropium or LABA, or both, as add‐on therapies to PDE₄ inhibitors (± ICS).

  • Effects of roflumilast on quality of life.

  • Better characterisation of the weight loss seen with PDE₄ inhibitors in COPD.

  • Better description of the nature of effects on exacerbations that do occur.

  • Use of PDE₄ inhibitors in acute exacerbations.

  • Cost‐effectiveness of PDE₄ inhibitors.

  • Increased exercise tolerance data for roflumilast.

  • Increased data on tetomilast.

  • Whether there is any benefit on cardiovascular outcomes for PDE₄ inhibitors in COPD.

  • Use of effects of PDE₄ inhibitors to better understand the pathophysiology of COPD.

  • Further evaluation of roflumilast 250 µg versus 500 µg daily.

  • Subgroup analysis of participants based on their weight.

  • Use of CAT score as an outcome.

  • Responder analyses (e.g. proportion of participants achieving a minimum clinically important difference).

What's new

Date Event Description
9 March 2020 New citation required but conclusions have not changed The 2020 update of this review includes 4 new trials of roflumilast ‐ Kavitha 2018; Liu 2018; RO‐2455‐402‐RD (ROBERT); Urban 2018 (ELASTIC) ‐ and 1 new trial of tetomilast ‐ NCT00874497 (EMPHASIS). Two new review authors (SJ and RF) were added, and 2 review authors (JC and BL) stepped down
9 March 2020 New search has been performed Literature search was run

History

Protocol first published: Issue 4, 2000
Review first published: Issue 5, 2011

Date Event Description
11 October 2016 New search has been performed New literature search was run
11 October 2016 New citation required but conclusions have not changed Five new eligible studies of roflumilast 500 μg were included ‐ RO‐2455‐301‐RD (ACROSS); RO‐2455‐404‐RD (REACT); Roflumilast DAL‐MD‐01; Roflumilast FLUI‐2011‐77; Roflumilast ROF‐MD‐07(RE2SPOND). No substantive changes were made to review findings
17 December 2013 Amended Typo in plain language summary title was amended
4 November 2013 Amended Risk of bias for Cilomilast 076 was added
6 June 2013 New search has been performed New literature search was run
6 June 2013 New citation required and conclusions have changed We included 7 new studies in this update and excluded 1 cross‐over trial. FDA report on psychiatric adverse events and suicides was included
Text was revised to take account of Cochrane reporting standards
'Summary of findings' table was added

Acknowledgements

The Background and Methods sections of this review are based on a standard template used by Cochrane Airways.

This review is dedicated to Professor Peter Black (deceased January 2010), who led development of the protocol and the early part of the review. Peter made significant contributions through research, teaching, and clinical practice to furthering of evidence‐based management of airways diseases.

We thank Claire Arandjus for her contribution to protocol development.

We thank Jimmy Chong for his contributions to the previous update of this review.

We thank Bonnie Leung for her contributions to the previous update of this review.

We thank Professor Milo Puhan for assistance in locating reports on the FDA website.

We thank Nycomed and Forest Laboratories for confirming some study details and results extracted from published articles and abstracts.

We thank GlaxoSmithKline (GSK) for study summaries available via the GSK online clinical study register.

The review authors and the Cochrane Airways editorial team are grateful to the following peer reviewers for their time and comments.

  • Dr PW Ind, Adjunct Reader NHLI, Hon Consultant Respiratory Physician, Imperial College NHS Healthcare Trust, UK.

  • Professor Peter Calverley, Institute of Ageing and Chronic Disease, University of Liverpool, UK.

  • Sarah Hodgkinson, Cochrane Central Executive Team.

This update was funded by the National Institute for Health Research Systematic Reviews Programme (project number 16/114/21). This project was also supported by the National Institute for Health Research (NIHR) via Cochrane Infrastructure funding to the Cochrane Airways Group. The views and opinions expressed therein are those of the authors and do not necessarily reflect those of the Health Research Systematic Reviews Programme, NIHR, NHS, or the Department of Health.

Appendices

Appendix 1. Sources and search methods for the Cochrane Airways Trials Register

Electronic searches: core databases

Database Frequency of search
CENTRAL (The Cochrane Library) Monthly
MEDLINE (Ovid SP) ALL Weekly
Embase (Ovid SP) Weekly
PsycINFO (Ovid SP) Monthly
CINAHL (EBSCO) Monthly
AMED (EBSCO) Monthly

 

Handsearches: core respiratory conference abstracts

Conference Years searched
American Academy of Allergy, Asthma and Immunology (AAAAI) 2001 onwards
American Thoracic Society (ATS) 2001 onwards
Asia Pacific Society of Respirology (APSR) 2004 onwards
British Thoracic Society Winter Meeting (BTS) 2000 onwards
Chest Meeting 2003 onwards
European Respiratory Society (ERS) 1992, 1994, 2000 onwards
International Primary Care Respiratory Group Congress (IPCRG) 2002 onwards
Thoracic Society of Australia and New Zealand (TSANZ) 1999 onwards

 

MEDLINE search strategy used to identify trials for the Cochrane Airways Trials Register

COPD  search

1. Lung Diseases, Obstructive/

2. exp Pulmonary Disease, Chronic Obstructive/

3. emphysema$.mp.

4. (chronic$ adj3 bronchiti$).mp.

5. (obstruct$ adj3 (pulmonary or lung$ or airway$ or airflow$ or bronch$ or respirat$)).mp.

6. COPD.mp.

7. COAD.mp.

8. COBD.mp.

9. AECB.mp.

10. or/1‐9

Filter to identify RCTs

1. exp "clinical trial [publication type]"/

2. (randomised or randomised).ab,ti.

3. placebo.ab,ti.

4. dt.fs.

5. randomly.ab,ti.

6. trial.ab,ti.

7. groups.ab,ti.

8. or/1‐7

9. Animals/

10. Humans/

11. 9 not (9 and 10)

12. 8 not 11

The MEDLINE strategy and RCT filter are adapted to identify trials in other electronic databases

Appendix 2. Search strategy to identify relevant trials from the Cochrane Airways Trials Register

#1 MeSH DESCRIPTOR Pulmonary Disease, Chronic Obstructive Explode All
#2 MeSH DESCRIPTOR Bronchitis, Chronic
#3 (obstruct*) near3 (pulmonary or lung* or airway* or airflow* or bronch* or respirat*)
#4 COPD:MISC1
#5 (COPD OR COAD OR COBD):TI,AB,KW
#6 #1 OR #2 OR #3 OR #4 OR #5
#7 MeSH DESCRIPTOR Phosphodiesterase 4 Inhibitors
#8 Phosphodiesterase*
#9 PDE4*
#10 roflumilast
#11 rolipram
#12 cilomilast
#13 ariflo
#14 SB207499
#15 Tetomilast
#16 ORIC485
#17 Oglemilast
#18 GRC‐3886
#19 QAK423
#20 Arofylline
#21 AWD12‐281
#22 #7 or #8 or #9 or #10 or #11 or #12 or #13 or #14 or #15 or #16 or #17 or #18 or #19 or #20 or #21
#23 #6 and #22

Appendix 3. Airways Group Trials Register search strategy (sensitive search)

PDE* or phosphodiesterase* or isoenzyme* or theophylline or rolipram or pentoxifylline or papaverine or milrinone or etazolate or etazolate or dyphylline or dipyridamole or caffeine or amrinone or aminophylline or isobutylxanthine or cilomilast or ariflo or cilostazol or enoximone or milrinone or olprinone or roflumilast or sb207499 or zardaverine or cilostamide or enoximone or trequinsin or Telomilast or IC485 or Oglemilast or QAK423 or GRC‐3886 or Arofylline or AWD12‐281

Data and analyses

Comparison 1. PDE4 inhibitor versus placebo (2020 update).

Outcome or subgroup title No. of studies No. of participants Statistical method Effect size
1.1 FEV₁ (by drug) 29 20815 Mean Difference (IV, Fixed, 95% CI) 49.33 [44.17, 54.49]
1.1.1 Tetomilast 50 µg 1 76 Mean Difference (IV, Fixed, 95% CI) 82.00 [‐50.84, 214.84]
1.1.2 Roflumilast 500 μg 18 14384 Mean Difference (IV, Fixed, 95% CI) 55.18 [48.65, 61.71]
1.1.3 Roflumilast 250 μg 3 1033 Mean Difference (IV, Fixed, 95% CI) 56.88 [24.38, 89.38]
1.1.4 Cilomilast 15 mg 10 5322 Mean Difference (IV, Fixed, 95% CI) 38.15 [29.41, 46.90]
1.2 FVC 17 22108 Mean Difference (IV, Fixed, 95% CI) 86.98 [74.65, 99.31]
1.3 PEF 5 4245 Mean Difference (IV, Fixed, 95% CI) 6.54 [3.95, 9.13]
1.3.1 Roflumilast 500 μg 4 3685 Mean Difference (IV, Fixed, 95% CI) 5.46 [2.74, 8.17]
1.3.2 Roflumilast 250 μg 1 347 Mean Difference (IV, Fixed, 95% CI) 7.00 [‐4.05, 18.05]
1.3.3 Cilomilast 15 mg 1 213 Mean Difference (IV, Fixed, 95% CI) 34.00 [20.14, 47.86]
1.4 SGRQ total score 11 7645 Mean Difference (IV, Fixed, 95% CI) ‐1.06 [‐1.68, ‐0.43]
1.4.1 Roflumilast 500 μg 2 722 Mean Difference (IV, Fixed, 95% CI) ‐1.87 [‐3.80, 0.06]
1.4.2 Roflumilast 250 μg 2 2229 Mean Difference (IV, Fixed, 95% CI) ‐0.64 [‐2.02, 0.74]
1.4.3 Cilomilast 15 mg 8 4694 Mean Difference (IV, Fixed, 95% CI) ‐1.06 [‐1.81, ‐0.31]
1.5 SGRQ symptom score 2 1048 Mean Difference (IV, Fixed, 95% CI) ‐1.53 [‐4.11, 1.06]
1.5.1 Roflumilast 1 835 Mean Difference (IV, Fixed, 95% CI) ‐1.00 [‐3.78, 1.78]
1.5.2 Cilomilast 1 213 Mean Difference (IV, Fixed, 95% CI) ‐4.80 [‐11.73, 2.13]
1.6 Number of participants with 1 or more exacerbations (by drug) 27 20382 Odds Ratio (M‐H, Fixed, 95% CI) 0.78 [0.73, 0.84]
1.6.1 Roflumilast 500 μg 16 14778 Odds Ratio (M‐H, Fixed, 95% CI) 0.79 [0.73, 0.86]
1.6.2 Cilomilast 10 5528 Odds Ratio (M‐H, Fixed, 95% CI) 0.76 [0.67, 0.85]
1.6.3 Tetomilast 50 µg 1 76 Odds Ratio (M‐H, Fixed, 95% CI) 2.45 [0.26, 23.13]
1.7 Exacerbation rate (inverse variance) 9   Rate Ratio (IV, Fixed, 95% CI) 0.88 [0.83, 0.93]
1.7.1 Roflumilast 8   Rate Ratio (IV, Fixed, 95% CI) 0.87 [0.82, 0.92]
1.7.2 Cilomilast 1   Rate Ratio (IV, Fixed, 95% CI) 0.95 [0.78, 1.17]
1.8 Borg Scale 6 2860 Mean Difference (IV, Fixed, 95% CI) ‐0.19 [‐0.33, ‐0.05]
1.8.1 Cilomilast 6 2860 Mean Difference (IV, Fixed, 95% CI) ‐0.19 [‐0.33, ‐0.05]
1.9 Shortness of Breath Questionnaire 2 1633 Mean Difference (IV, Fixed, 95% CI) ‐1.09 [‐2.47, 0.28]
1.10 Summary symptom score 5 6186 Std. Mean Difference (IV, Fixed, 95% CI) ‐0.02 [‐0.07, 0.03]
1.10.1 Roflumilast 2 4287 Std. Mean Difference (IV, Fixed, 95% CI) ‐0.02 [‐0.08, 0.04]
1.10.2 Cilomilast 3 1899 Std. Mean Difference (IV, Fixed, 95% CI) ‐0.04 [‐0.13, 0.06]
1.11 Breathlessness Cough and Sputum Scale (BCSS) (tetomilast 50 µg) 1   Mean Difference (IV, Fixed, 95% CI) Subtotals only
1.11.1 Breathlessness 1 22 Mean Difference (IV, Fixed, 95% CI) ‐0.07 [‐0.77, 0.63]
1.11.2 Cough 1 22 Mean Difference (IV, Fixed, 95% CI) 0.23 [‐0.54, 1.00]
1.11.3 Sputum 1 22 Mean Difference (IV, Fixed, 95% CI) ‐0.16 [‐0.97, 0.65]
1.12 6‐minute walk test 6 2055 Mean Difference (IV, Fixed, 95% CI) 3.50 [‐5.84, 12.85]
1.12.1 Roflumilast 2 107 Mean Difference (IV, Fixed, 95% CI) 52.61 [‐0.21, 105.42]
1.12.2 Cilomilast 4 1948 Mean Difference (IV, Fixed, 95% CI) 1.92 [‐7.58, 11.41]
1.13 Number of participants experiencing an adverse event 30 21310 Odds Ratio (M‐H, Fixed, 95% CI) 1.30 [1.22, 1.38]
1.13.1 Roflumilast 500 μg 15 14684 Odds Ratio (M‐H, Fixed, 95% CI) 1.34 [1.24, 1.43]
1.13.2 Cilomilast 15 mg 14 6542 Odds Ratio (M‐H, Fixed, 95% CI) 1.21 [1.08, 1.36]
1.13.3 Tetomilast 50 µg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 0.62 [0.25, 1.57]
1.14 Number of participants experiencing an adverse event (roflumilast 500 μg vs 250 μg) 4 1977 Odds Ratio (M‐H, Fixed, 95% CI) 1.21 [1.01, 1.46]
1.15 Diarrhoea 29 20623 Odds Ratio (M‐H, Fixed, 95% CI) 3.10 [2.74, 3.50]
1.15.1 Roflumilast 14 13997 Odds Ratio (M‐H, Fixed, 95% CI) 3.65 [3.10, 4.28]
1.15.2 Cilomilast 14 6542 Odds Ratio (M‐H, Fixed, 95% CI) 2.47 [2.05, 2.98]
1.15.3 Tetomilast 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 1.68 [0.31, 9.24]
1.16 Nausea 27 20949 Odds Ratio (M‐H, Fixed, 95% CI) 3.79 [3.24, 4.43]
1.16.1 Roflumilast 500 μg 12 13467 Odds Ratio (M‐H, Fixed, 95% CI) 3.25 [2.60, 4.07]
1.16.2 Roflumilast 250 μg 1 856 Odds Ratio (M‐H, Fixed, 95% CI) 3.97 [0.91, 17.39]
1.16.3 Cilomilast 15 mg 14 6542 Odds Ratio (M‐H, Fixed, 95% CI) 4.37 [3.49, 5.47]
1.16.4 Tetomilast 50 µg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 2.00 [0.20, 20.09]
1.17 Vomiting 12 5986 Odds Ratio (M‐H, Fixed, 95% CI) 3.95 [2.78, 5.60]
1.17.1 Roflumilast 2 993 Odds Ratio (M‐H, Fixed, 95% CI) 2.32 [0.53, 10.23]
1.17.2 Cilomilast 10 4993 Odds Ratio (M‐H, Fixed, 95% CI) 4.06 [2.83, 5.82]
1.18 Dyspepsia 13 6247 Odds Ratio (M‐H, Fixed, 95% CI) 3.17 [2.33, 4.30]
1.18.1 Roflumilast 1 626 Odds Ratio (M‐H, Fixed, 95% CI) 7.07 [0.36, 137.40]
1.18.2 Cilomilast 12 5621 Odds Ratio (M‐H, Fixed, 95% CI) 3.13 [2.30, 4.27]
1.19 Weight loss 12 12462 Odds Ratio (M‐H, Fixed, 95% CI) 3.72 [3.09, 4.47]
1.19.1 Roflumilast 11 12378 Odds Ratio (M‐H, Fixed, 95% CI) 3.80 [3.15, 4.58]
1.19.2 Tetomilast 50 µg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 0.12 [0.01, 2.63]
1.20 Withdrawals due to adverse events 31 21358 Odds Ratio (M‐H, Fixed, 95% CI) 1.89 [1.73, 2.07]
1.20.1 Roflumilast 500 μg 16 14729 Odds Ratio (M‐H, Fixed, 95% CI) 1.90 [1.70, 2.12]
1.20.2 Cilomilast 15 mg 14 6545 Odds Ratio (M‐H, Fixed, 95% CI) 1.90 [1.61, 2.24]
1.20.3 Tetomilast 50 mg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 0.79 [0.20, 3.18]
1.21 Headache 23 19215 Odds Ratio (M‐H, Fixed, 95% CI) 1.69 [1.46, 1.94]
1.21.1 Roflumilast 500 μg 12 13565 Odds Ratio (M‐H, Fixed, 95% CI) 2.13 [1.74, 2.59]
1.21.2 Roflumilast 250 μg 1 347 Odds Ratio (M‐H, Fixed, 95% CI) 0.98 [0.24, 3.99]
1.21.3 Cilomilast 15 mg 11 5303 Odds Ratio (M‐H, Fixed, 95% CI) 1.32 [1.08, 1.62]
1.22 Abdominal pain 15 8329 Odds Ratio (M‐H, Fixed, 95% CI) 2.02 [1.62, 2.52]
1.22.1 Roflumilast 3 2641 Odds Ratio (M‐H, Fixed, 95% CI) 2.77 [1.38, 5.56]
1.22.2 Cilomilast 11 5604 Odds Ratio (M‐H, Fixed, 95% CI) 1.97 [1.55, 2.49]
1.22.3 Tetomilast 50 µg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 0.97 [0.15, 6.13]
1.23 Influenza‐like symptoms 9 11460 Odds Ratio (M‐H, Fixed, 95% CI) 1.09 [0.87, 1.36]
1.23.1 Roflumilast 500 μg 7 10147 Odds Ratio (M‐H, Fixed, 95% CI) 1.11 [0.87, 1.41]
1.23.2 Roflumilast 250 μg 1 347 Odds Ratio (M‐H, Fixed, 95% CI) 1.98 [0.18, 22.00]
1.23.3 Cilomilast 15 mg 2 966 Odds Ratio (M‐H, Fixed, 95% CI) 0.88 [0.44, 1.75]
1.24 Upper respiratory tract infection 21 17022 Odds Ratio (M‐H, Fixed, 95% CI) 0.91 [0.81, 1.04]
1.24.1 Roflumilast 500 μg 11 11539 Odds Ratio (M‐H, Fixed, 95% CI) 0.92 [0.77, 1.09]
1.24.2 Roflumilast 250 μg 2 1203 Odds Ratio (M‐H, Fixed, 95% CI) 0.84 [0.54, 1.31]
1.24.3 Cilomilast 15 mg 10 4280 Odds Ratio (M‐H, Fixed, 95% CI) 0.92 [0.75, 1.13]
1.25 Psychiatric adverse events (roflumilast) 1   Odds Ratio (M‐H, Fixed, 95% CI) Subtotals only
1.25.1 Roflumilast 500 μg 1 11168 Odds Ratio (M‐H, Fixed, 95% CI) 2.13 [1.79, 2.54]
1.25.2 Roflumilast 250 μg 1 6288 Odds Ratio (M‐H, Fixed, 95% CI) 0.87 [0.56, 1.33]
1.26 Anxiety or anxiety disorder (roflumilast) 1   Odds Ratio (M‐H, Fixed, 95% CI) Subtotals only
1.26.1 Roflumilast 500 μg 1 11168 Odds Ratio (M‐H, Fixed, 95% CI) 1.81 [1.26, 2.62]
1.26.2 Roflumilast 250 μg 1 6288 Odds Ratio (M‐H, Fixed, 95% CI) 0.94 [0.40, 2.21]
1.27 Depression (roflumilast) 1   Odds Ratio (M‐H, Fixed, 95% CI) Subtotals only
1.27.1 Roflumilast 500 μg 1 11168 Odds Ratio (M‐H, Fixed, 95% CI) 1.59 [1.11, 2.27]
1.27.2 Roflumilast 250 μg 1 6288 Odds Ratio (M‐H, Fixed, 95% CI) 0.56 [0.20, 1.56]
1.28 Insomnia and sleep disorders (roflumilast) 4   Odds Ratio (M‐H, Fixed, 95% CI) Subtotals only
1.28.1 Roflumilast 500 μg 4 15482 Odds Ratio (M‐H, Fixed, 95% CI) 2.67 [2.11, 3.38]
1.28.2 Roflumilast 250 μg 1 6288 Odds Ratio (M‐H, Fixed, 95% CI) 1.48 [0.81, 2.70]
1.29 Serious adverse events 29 19191 Odds Ratio (M‐H, Fixed, 95% CI) 0.99 [0.91, 1.07]
1.29.1 Roflumilast 500 μg 14 12562 Odds Ratio (M‐H, Fixed, 95% CI) 1.02 [0.93, 1.12]
1.29.2 Cilomilast 15 mg 14 6545 Odds Ratio (M‐H, Fixed, 95% CI) 0.87 [0.72, 1.06]
1.29.3 Tetomilast 50 µg 1 84 Odds Ratio (M‐H, Fixed, 95% CI) 0.60 [0.23, 1.55]
1.30 Mortality 27 19786 Odds Ratio (M‐H, Fixed, 95% CI) 0.98 [0.77, 1.24]
1.30.1 Roflumilast 13 13370 Odds Ratio (M‐H, Fixed, 95% CI) 1.01 [0.79, 1.30]
1.30.2 Cilomilast 13 6332 Odds Ratio (M‐H, Fixed, 95% CI) 0.70 [0.34, 1.45]
1.30.3 Tetomilast 1 84 Odds Ratio (M‐H, Fixed, 95% CI) Not estimable
1.31 FEV₁ (by mean COPD severity) 22 16813 Mean Difference (IV, Fixed, 95% CI) 52.78 [46.73, 58.83]
1.31.1 GOLD grade I + II (FEV₁ ≥ 50% predicted) 10 4801 Mean Difference (IV, Fixed, 95% CI) 51.82 [39.03, 64.60]
1.31.2 GOLD grade III + IV (FEV₁ < 50% predicted) 12 12012 Mean Difference (IV, Fixed, 95% CI) 53.06 [46.19, 59.92]
1.32 FEV₁ (roflumilast 500 μg vs 250 μg) 3 1560 Mean Difference (IV, Fixed, 95% CI) 22.61 [‐5.95, 51.16]
1.32.1 Roflumilast 250 ug 3 1560 Mean Difference (IV, Fixed, 95% CI) 22.61 [‐5.95, 51.16]
1.33 FEV₁ (by study duration) 28 19939 Mean Difference (IV, Fixed, 95% CI) 49.09 [43.86, 54.32]
1.33.1 Duration ≤ 12 weeks 8 1191 Mean Difference (IV, Fixed, 95% CI) 101.71 [70.96, 132.46]
1.33.2 Duration 24 to 26 weeks 13 8086 Mean Difference (IV, Fixed, 95% CI) 46.14 [38.44, 53.84]
1.33.3 Duration 52 weeks 7 10662 Mean Difference (IV, Fixed, 95% CI) 48.77 [41.44, 56.10]
1.34 FEV₁ (additional medication) 28 19719 Mean Difference (IV, Fixed, 95% CI) 49.08 [43.85, 54.31]
1.34.1 Long‐acting bronchodilator 2 1645 Mean Difference (IV, Fixed, 95% CI) 60.52 [40.57, 80.46]
1.34.2 Corticosteroids 3 2904 Mean Difference (IV, Fixed, 95% CI) 42.26 [25.46, 59.05]
1.34.3 PDE₄i treatment only 20 10323 Mean Difference (IV, Fixed, 95% CI) 44.80 [37.69, 51.91]
1.34.4 Various concomitant treatments 3 4847 Mean Difference (IV, Fixed, 95% CI) 56.58 [46.91, 66.25]
1.35 FEV₁ (random‐effects model) 29 20015 Mean Difference (IV, Random, 95% CI) 51.49 [42.87, 60.10]
1.36 FEV₁ (published vs unpublished) 29 20015 Mean Difference (IV, Fixed, 95% CI) 49.28 [44.05, 54.51]
1.36.1 Published 20 15398 Mean Difference (IV, Fixed, 95% CI) 55.75 [49.45, 62.06]
1.36.2 Unpublished 9 4617 Mean Difference (IV, Fixed, 95% CI) 35.05 [25.70, 44.40]
1.37 SGRQ total score (by mean COPD severity) 8 4851 Mean Difference (IV, Fixed, 95% CI) ‐1.56 [‐2.39, ‐0.74]
1.37.1 GOLD grade I + II 3 2042 Mean Difference (IV, Fixed, 95% CI) ‐1.62 [‐2.80, ‐0.44]
1.37.2 GOLD grade III + IV 5 2809 Mean Difference (IV, Fixed, 95% CI) ‐1.51 [‐2.67, ‐0.34]
1.38 SGRQ total score (by duration) 11 7069 Mean Difference (IV, Fixed, 95% CI) ‐0.99 [‐1.65, ‐0.33]
1.38.1 Duration < 12 weeks 2 240 Mean Difference (IV, Fixed, 95% CI) ‐4.19 [‐7.60, ‐0.78]
1.38.2 Duration 24 to 26 weeks 7 4600 Mean Difference (IV, Fixed, 95% CI) ‐1.18 [‐1.94, ‐0.42]
1.38.3 Duration 52 weeks 2 2229 Mean Difference (IV, Fixed, 95% CI) 0.26 [‐1.18, 1.69]
1.39 SGRQ total score (by published vs unpublished) 11 7069 Mean Difference (IV, Fixed, 95% CI) ‐1.00 [‐1.65, ‐0.34]
1.39.1 Published 5 3079 Mean Difference (IV, Fixed, 95% CI) ‐1.98 [‐3.07, ‐0.89]
1.39.2 Unpublished 6 3990 Mean Difference (IV, Fixed, 95% CI) ‐0.43 [‐1.26, 0.40]
1.40 Number of participants on roflumilast with 1 or more exacerbations (additional medication) 15 14698 Odds Ratio (M‐H, Fixed, 95% CI) 0.79 [0.73, 0.85]
1.40.1 Long‐acting bronchodilators 3 1834 Odds Ratio (M‐H, Fixed, 95% CI) 0.69 [0.54, 0.88]
1.40.2 Corticosteroids 1 2686 Odds Ratio (M‐H, Fixed, 95% CI) 0.81 [0.70, 0.95]
1.40.3 Treatment only 7 5145 Odds Ratio (M‐H, Fixed, 95% CI) 0.79 [0.67, 0.93]
1.40.4 Various concomitant treatments 4 5033 Odds Ratio (M‐H, Fixed, 95% CI) 0.81 [0.72, 0.91]
1.41 FVC ML (roflumilast 500 µg, endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.42 FEV₁ (by unknown COPD severity) 1 76 Mean Difference (IV, Fixed, 95% CI) 82.00 [‐50.84, 214.84]
1.43 FEV₁ (by duration, endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.44 FEV₁ (random‐effects model, endpoint data) 1 60 Mean Difference (IV, Random, 95% CI) 0.43 [0.31, 0.55]
1.45 FEV₁ (by moderate to severe COPD severity, endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.46 FEV₁ (roflumilast 500 µg, endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.43 [0.31, 0.55]
1.47 FEV₁ ML (additional medication (PDE₄i only) endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.48 FEV₁ (published, endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.49 FEV₁ (roflumilast 500 μg by mean COPD severity) 16 13896 Mean Difference (IV, Fixed, 95% CI) 55.51 [48.88, 62.14]
1.49.1 GOLD grade I + II (FEV₁ ≥ 50% predicted) 7 3341 Mean Difference (IV, Fixed, 95% CI) 69.83 [53.34, 86.33]
1.49.2 GOLD grade III + IV (FEV₁ < 50% predicted) 9 10555 Mean Difference (IV, Fixed, 95% CI) 52.75 [45.52, 59.99]
1.50 FEV₁ (unknown additional medication) 1 76 Mean Difference (IV, Fixed, 95% CI) 82.00 [‐50.84, 214.84]
1.51 FEV₁ (by moderate to severe COPD severity, roflumilast 500 µg endpoint) 1 60 Mean Difference (IV, Fixed, 95% CI) 0.52 [0.25, 0.79]
1.52 FEV₁ (by unknown COPD severity, roflumilast 500 µg ) 1 76 Mean Difference (IV, Fixed, 95% CI) 82.00 [‐50.84, 214.84]

1.41. Analysis.

1.41

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 41: FVC ML (roflumilast 500 µg, endpoint)

1.42. Analysis.

1.42

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 42: FEV₁ (by unknown COPD severity)

1.43. Analysis.

1.43

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 43: FEV₁ (by duration, endpoint)

1.44. Analysis.

1.44

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 44: FEV₁ (random‐effects model, endpoint data)

1.45. Analysis.

1.45

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 45: FEV₁ (by moderate to severe COPD severity, endpoint)

1.46. Analysis.

1.46

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 46: FEV₁ (roflumilast 500 µg, endpoint)

1.47. Analysis.

1.47

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 47: FEV₁ ML (additional medication (PDE₄i only) endpoint)

1.48. Analysis.

1.48

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 48: FEV₁ (published, endpoint)

1.49. Analysis.

1.49

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 49: FEV₁ (roflumilast 500 μg by mean COPD severity)

1.50. Analysis.

1.50

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 50: FEV₁ (unknown additional medication)

1.51. Analysis.

1.51

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 51: FEV₁ (by moderate to severe COPD severity, roflumilast 500 µg endpoint)

1.52. Analysis.

1.52

Comparison 1: PDE4 inhibitor versus placebo (2020 update), Outcome 52: FEV₁ (by unknown COPD severity, roflumilast 500 µg )

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Cilomilast 039.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 102 centres in Canada, Mexico, and the USA
Participants: 647 (15 mg cilomilast: 431, placebo: 216)
Baseline characteristics: mean age 65 years, 62% male, mean FEV₁ % predicted 49.7%, mean smoking history 59.9 pack‐years for cilomilast and 56.1 pack‐years for placebo, or current smokers (44% and 47%, respectively)
Inclusion criteria: FEV₁/FVC ≤ 0.7, FEV₁ 30% to 70% with smoking history > 10 pack‐years or current smokers
Exclusion criteria: active tuberculosis, lung cancer, bronchiectasis
Total numbers of participant withdrawals: 137 (32%) and 52 (24%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind. Placebo tablets to assess suitability
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: "the only other permitted medications for the treatment of airways disease were stable doses of Ipratropium, via a metered‐dose inhaler, and mucolytic agents"

  • SABA: "...the short‐acting β₂‐agonist albuterol, which was administered via a metered‐dose inhaler, was supplied for the relief of acute respiratory symptoms"

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: lung function; change in FEV₁; SGRQ averaged over 24 weeks
Secondary outcomes: incidence rate of COPD exacerbations; adverse events; FVC at trough; 6MWT; post‐exercise dyspnoea
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that the randomisation process was adequate due to pharma sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk "The primary reasons for the withdrawal of subjects from the study prior to randomisation were the failure to meet inclusion/exclusion criteria (15.4%) and the presence of adverse effects, including COPD exacerbations (8.5%). More subjects receiving cilomilast than placebo withdrew from the double‐blind phase of study (31.8% (n = 137) versus 24.1% (n = 52)"
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website
Other bias Low risk Baseline anticholinergic, beta₂‐agonist, or corticosteroid use 54% in cilomilast, 58% placebo used ipratropium; 99% in cilomilast, 100% placebo used albuterol; 9% in cilomilast, 12% placebo used salmeterol; 7% in cilomilast, 8% placebo used triamcinolone; 6% in cilomilast, 7% placebo used beclomethasone

Cilomilast 042.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 98 centres in Australia and New Zealand, Germany, Spain, South Africa, and the UK
Participants: 700 (15 mg cilomilast: 474, placebo: 226)
Baseline characteristics: mean age 64.6 years, 80% male, mean FEV₁ % predicted 49% with 5.1% reversibility, DLCO 71% predicted, also with higher rates of chronic bronchitis 80.1%. 45% active smokers
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7, FEV₁ 30% to 70% with smoking history > 10 pack‐years
Exclusion criteria: active tuberculosis, lung cancer, bronchiectasis
Total numbers of participant withdrawals: 122 (26%) and 51 (23%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind with placebo
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: 2% in cilomilast, 3% placebo used salbutamol; 3% in cilomilast, 1% placebo used ipratropium

  • SABA: "albuterol MDI was used as rescue medication"

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: lung function; change in FEV₁; SGRQ averaged over 24 weeks
Secondary outcomes: incidence rate of COPD exacerbations; summary symptom score; FVC at trough; 6MWT; post‐exercise dyspnoea
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that the randomisation method was adequate due to pharma sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 51 (23%) placebo, 122 (26%) cilomilast, primarily due to adverse events, of which most were not from COPD exacerbations
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 076.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Analysis was done on per‐protocol population
Participants Setting: not stated
Participants: 59 (15 mg cilomilast: 29, placebo: 30)
Baseline characteristics: mean age 61 to 62 years, 81% male, 53% active smokers, mean 46 pack‐years, 53% to 58% FEV₁ predicted
Inclusion criteria: aged 40 to 80 years, fixed airflow obstruction, smoking history > 10 pack‐years
Exclusion criteria: not stated
Total numbers of participant withdrawals: 4 (14%) and 2 (7%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind with placebo
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: "14 of 59 used ipratropium bromide at a constant dosage (8 in the placebo group, 6 in the cilomilast group)"

  • SABA: "all patients were given albuterol for use as required"

  • Corticosteroid: none

  • LABA: none


Used alongside SABA (available to all) and anticholingeric drugs (offered to 24%)
Outcomes Primary outcome: change in neutrophil percentage in induced sputum
Secondary outcomes: FEV₁; numbers of subepithelial CD8+ cells, CD 68+ cells, epithelial, and subepithelial neutrophils
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that the randomisation process was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk "One patient was lost to follow‐up 3 days after randomisation and another withdrawn for non‐compliance 32 days after randomisation. Four patients were withdrawn after adverse events"
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 091.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 110 centres in Belgium, Finland, France, Italy, the Netherlands, Norway, Portugal, Spain, and the UK
Participants: 711 (15 mg cilomilast: 469, placebo: 242)
Baseline characteristics: mean age 64.6 years, 86% male, mean FEV₁ % predicted 53% with 5.0% reversibility, 38% active smokers
Inclusion criteria: FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years
Exclusion criteria: active tuberculosis, lung cancer, bronchiectasis
Total numbers of participant withdrawals: 121 (26%) and 63 (26%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind with placebo
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: 0.9% in cilomilast, 4% placebo used salbutamol; 1% in cilomilast, 3% placebo used ipratropium

  • SABA: "albuterol MDI was used as rescue medication"

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: lung function; change in FEV₁; SGRQ averaged over 24 weeks
Secondary outcomes: incidence rate of COPD exacerbations; summary symptom score; FVC at trough; 6MWT; post‐exercise dyspnoea
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that the randomisation process was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 63 (26%) placebo, 121 (26%) cilomilast, primarily due to adverse events, of which most were not due to COPD exacerbations
Selective reporting (reporting bias) Unclear risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 103657.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 103 centres in the USA
Participants: 613 (15 mg cilomilast: 296, placebo: 317)
Baseline characteristics: mean age 63.2 years placebo, 63.1 years cilomilast, 47% male placebo, 46% male cilomilast. Mean FEV₁ % predicted not available
Inclusion criteria: aged ≥ 40 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, ≤ 70% post‐albuterol reversibility, ≤ 15% or ≤ 200 mL (or both) post‐albuterol FEV₁ ≤ 70% of predicted normal, ≥ 1 COPD exacerbation within 12 months before screening
Exclusion criteria: not stated
Total numbers of participant withdrawals: 105 (35%) and 76 (24%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcomes: change from baseline to endpoint in trough pre‐bronchodilator FEV₁; change in total SGRQ score averaged over 24 weeks
Secondary outcomes: changes from baseline in clinic trough FVC; time to first level 2 or level 3 COPD exacerbation
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk The trial was double‐blinded (participants and investigator). It is not clear if the investigator was administering the treatment
Blinding of outcome assessment (detection bias)
All outcomes Low risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 76 (24%) placebo, 105 (35%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. The trial was registered at clinicaltrials.gov
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 110.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Analysis was done on per‐protocol population
Participants Setting: 10 centres in the USA
Participants: 65 (15 mg cilomilast: 31, placebo: 34)
Baseline characteristics: mean age 64.4 years placebo and 66.1 years cilomilast, 67% male placebo and 84% male cilomilast, mean FEV₁ % predicted not available
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, post‐salbutamol reversibility ≤ 15% or 200 mL, post‐salbutamol FEV₁ ≥ 1.0 L and between 30% and 70% predicted
Exclusion criteria: not stated
Total numbers of participant withdrawals: 1 (3%) and 1 (3%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcome: change from baseline at endpoint in neutrophils as a percentage of total cells in induced sputum
Secondary outcomes: FVC at trough; sputum macrophages, eosinophils, and lymphocytes as a percentage of total cells in induced sputum; total cell counts in induced sputum
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical sponsorship
Allocation concealment (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Low risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 1 (3%) placebo, 1 (3%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 111.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 32 centres in the USA, Canada, and Australia
Participants: 156 (15 mg cilomilast: 79, placebo: 77)
Baseline characteristics: mean age 64.2 years placebo and 65 years cilomilast, 66% male placebo and 65% male cilomilast, mean FEV₁ % predicted not available
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, post‐salbutamol reversibility ≤ 15% or 200 mL, post‐salbutamol FEV₁ ≥ 1.0 L and between 30% and 70% predicted, baseline RV (from plethysmography) ≥ 120% predicted RV
Exclusion criteria: not stated
Total numbers of participant withdrawals: 15 (19%) and 14 (18%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcome: change from baseline to endpoint in volume of trapped gas (D)
Secondary outcomes: lung volume measurements, including SVC and RV; 6MWT; exertional dyspnoea
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical sponsorship
Allocation concealment (selection bias) Low risk Assumed that trialists used a robust method to carry out the allocation concealment process because of pharmaceutical sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 14 (18%) placebo, 15 (19%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 121.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 22 centres in China
Participants: 1018 (15 mg cilomilast: 678, placebo: 340)
Baseline characteristics: mean age 63.9 years placebo and 64.6 years cilomilast, 91% male placebo and 93% male cilomilast, mean FEV₁ % predicted not available
Inclusion criteria: aged 40 to 75 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, documented history of COPD exacerbations each year for 3 years before screening, ≥ 1 exacerbation in the last year that required oral corticosteroids or antibiotics, post‐salbutamol reversibility ≤ 15% or 200 mL, post‐salbutamol FEV₁ ≥ 1.0 L and between 25% and 70% predicted, % predicted FRC ≥ 120% from plethysmography
Exclusion criteria: not stated
Total numbers of participant withdrawals: 124 (18%) and 35 (10%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcome: change from baseline to endpoint in trough pre‐bronchodilator FEV₁
Secondary outcomes: time to first level 2 or level 3 COPD exacerbation (level 2 is defined as acute worsening of COPD that requires additional treatment or hospital outpatient visit; level 3 is hospital admission for treatment); change from baseline to endpoint RV and FRC; change from baseline total score on SGRQ
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that trialists used a robust method to carry out the randomisation process because of pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that trialists used a robust method to carry out the allocation concealment process because of pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 35 (10%) placebo, 124 (18%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 156.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 132 centres in USA and Canada
Participants: 825 (15 mg cilomilast: 418, placebo: 407)
Baseline characteristics: mean age 64.4 years placebo and 64.5 years cilomilast, 62% male placebo and 56% male cilomilast, > 50% predicted FEV₁ for both groups
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, post‐salbutamol reversibility ≤ 15% or 200 mL, post‐salbutamol FEV₁ ≥ 1.0 L and between 30% and 70% predicted
Exclusion criteria: not stated
Total numbers of participant withdrawals: 143 (34%) and 96 (24%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric 8.1% in cilomilast, 8.6% placebo used salbutamol; 1.7% in cilomilast, 2% placebo used ipratropium bromide

  • SABA: "albuterol MDI was used as rescue medication"

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: change from baseline to endpoint in trough pre‐bronchodilator FEV₁; change in total SGRQ score averaged over 24 weeks
Secondary outcomes: post‐exercise breathlessness; clinic trough FVC; time to first level 2 or level 3 COPD exacerbation
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk The randomisation code was provided by RAMOS (registration and medication ordering system)
Allocation concealment (selection bias) Low risk No further information on allocation concealment method
Blinding of participants and personnel (performance bias)
All outcomes Low risk Patients and personnel did not know which treatment had been allocated
Blinding of outcome assessment (detection bias)
All outcomes Low risk Outcome assessors did not know which treatment had been allocated
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 96 (24%) placebo, 143 (34%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 157.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 137 centres from 18 countries
Participants: 907 (15 mg cilomilast: 455, placebo: 452)
Baseline characteristics: mean age 63.3 years placebo and 64.6 years cilomilast, 73% male placebo and 78% male cilomilast, 42% current smokers
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, poor reversibility of airway obstruction defined by ≤ 10% predicted normal or ≤ 200 mL (or both) increase in FEV₁ after administration of salbutamol 400 µg via MDI at screening, post‐salbutamol FEV₁ between 30% and 70% predicted normal at screening
Exclusion criteria: not stated
Total numbers of participant withdrawals: 167 (37%) and 121 (27%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcomes: mean change from baseline in trough pre‐bronchodilator FEV₁ averaged over 52 weeks; incidence rate of level 2 (moderate) and level 3 (severe) COPD exacerbations during treatment period
Secondary outcomes: time to first level 2 or level 3 COPD exacerbation; quality of life determined by SGRQ
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Randomisation code was generated via the pharmaceutical company's coding memo system in blocks
Allocation concealment (selection bias) Low risk No further information on allocation concealment method
Blinding of participants and personnel (performance bias)
All outcomes Low risk Patients and investigator were blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 121 (27%) placebo, 167 (37%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 168.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: not stated
Participants Setting: 42 centres in the USA
Participants: 306 (15 mg cilomilast: 203, placebo: 103)
Baseline characteristics: mean age 64.3 years placebo and 65.0 years cilomilast, 64% male placebo and 70% male cilomilast
Inclusion criteria: pre‐albuterol FEV₁/FVC ≤ 0.7, post‐albuterol FEV₁ between 30% and 70% predicted
Exclusion criteria: not stated
Total numbers of participant withdrawals: 61 (30%) and 14 (14%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcome: no primary efficacy or safety analyses defined; descriptive statistics of change from baseline in minimum and maximum heart rate via 24‐hour Holter monitoring reported
Secondary outcome: no secondary efficacy or safety analyses defined
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk The randomisation sequence was generated by the pharmaceutical company's biometrics unit
Allocation concealment (selection bias) Unclear risk No further information on allocation concealment method
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 14 (14%) placebo, 61 (30%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 180.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 18 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 34 centres in the USA, Canada, and South America
Participants: 199 (15 mg cilomilast: 97, placebo: 102)
Baseline characteristics: mean age 64.7 years placebo and 63.7 years cilomilast, 76% male placebo and 69% male cilomilast
Inclusion criteria: age ≥ 40 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, baseline FEV₁ < 70% predicted normal, moderate to severe chronic dyspnoea defined by BDI focal score ≤ 7, evidence of hyperinflation defined by RFRC ≥ 140% predicted, exercise limitation defined as peak symptom limited VO₂ < 75%
Exclusion criteria: not stated
Total numbers of participant withdrawals: 24 (25%) and 13 (13%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcome: change from baseline at endpoint in RFRC
Secondary outcomes: change from baseline at endpoint in IC during exercise; exertional dyspnoea as measured by the modified Borg Scale
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Patients were randomised via a call to the sponsor's medication ordering system, during which the patient's subject number was confirmed and the patient was provided with a 6‐digit container number for identification of the initial bottle of double‐blind medication
Allocation concealment (selection bias) Unclear risk No further information on allocation concealment method.
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double blind. Cilomilast and matched placebo tablets were identical in appearance, and only the double‐blind medication included the container number
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 13 (13%) placebo, 24 (25%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Cilomilast 181.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 13 weeks
Analysis was done on the per‐protocol population
Participants Setting: 27 centres in Australia, Canada, Finland, Ireland, Lithuania, Norway, Romania, Slovakia, Slovenia, South Africa, Sweden, and the UK
Participants: 127 (15 mg cilomilast: 65, placebo: 62)
Baseline characteristics: mean age 63.4 years placebo and 61.4 years cilomilast, 76% male placebo and 72% male cilomilast
Inclusion criteria: aged 40 to 80 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, post‐bronchodilator FEV₁ between 40% and 80% predicted normal, poor reversibility of ≤ 10% or 200 mL increase in FEV₁
Exclusion criteria: not stated
Total numbers of participant withdrawals: 8 (12%) and 6 (10%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Cilomilast 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: no information available

  • SABA: no information available

  • Corticosteroid: no information available

  • LABA: no information available

Outcomes Primary outcomes: change from baseline at endpoint in CD68+ (macrophages) and CD8+ (cytotoxic T lymphocytes) per unit area of tissue
Secondary outcomes: change from baseline in numbers of subepithelial cells per unit area in biopsy for neutrophil elastase‐positive (ne+) cells, CD4+, IL‐8 mRNA‐positive cells, TNF‐alpha mRNA‐positive cells
Notes Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk A central randomisation schedule that was balanced at site level. An interactive voice response system was used to generate a random number to assign eligible participants
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind. Participants and personnel were blind to which treatment they were assigned to. Cilomilast and matched placebo tablets were identical in appearance, and only the double‐blind medication included the container number
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants withdrawn 6 (10%) placebo, 8 (12%) cilomilast
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website only
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Compton 2001.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 6 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 60 centres in Austria, France, Germany, the Netherlands, and the UK
Participants: 424 (5 mg cilomilast: 109, 10 mg cilomilast: 102, 15 mg cilomilast: 107, placebo: 106)
Baseline characteristics: mean age 62 to 63 years, 75% to 78% male, mean FEV₁ % predicted 46.8%, mean smoking history 36 to 43 (SD 22.4) pack‐years
Inclusion criteria: FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years
Exclusion criteria: asthma, poorly controlled COPD needing hospital visit 6 weeks before study, recent COPD exacerbations, recent corticosteroid use
Total numbers of participant withdrawals: 18 (17%) and 17 (16%) from treatment and control groups, respectively
Interventions Run‐in: 2 weeks, single‐blind, placebo tablets to assess compliance
  • Cilomilast 5 mg, 10 mg, 15 mg twice daily

  • Placebo twice daily


Concomitant medication
  • Short‐acting anticholingeric: 382 (90%) participants were given concomitant treatment for COPD during the study; 267 (70%) salbutamol and 115 (30%) ipratropium bromide

  • SABA: salbutamol used in 70%

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: lung function: change in FEV₁; SGRQ
Secondary outcomes: peak expiratory flow and FVC; first dose effect of active treatment on FEV₁
Notes Post‐bronchodilator results not given, so pre‐bronchodilator values used in analysis. Funded by GlaxoSmithKline
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Randomised. Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded. Participants were not aware of which treatment they were receiving because cilomilast and matched placebo tablets were identical in appearance
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk The trial was double‐blinded, but it is unclear who assessed the outcomes
Incomplete outcome data (attrition bias)
All outcomes Low risk "14 patients (13%) taking cilomilast 15 mg had adverse events leading to patient withdrawal, as did 12 each in the 5 and 10 mg groups (11 and 12%, respectively) and eight (8%) in the placebo group"
Selective reporting (reporting bias) Unclear risk Unclear whether outcomes were assessed as planned; it was not possible to find the trial in the GSK registry
Other bias Low risk 102 (24%) participants had been taking long‐acting β₂‐agonists (e.g. salmeterol, formoterol). 331 (78%) individuals had taken other medications for their COPD, the most common being inhaled steroids; 229 (54%) took beclomethasone, budesonide, or fluticasone

COPD safety pool.

Study characteristics
Methods 14 double‐blind and placebo‐controlled studies (Roflumilast FK1 101; Roflumilast FK1 103; Roflumilast IN‐108; Roflumilast M2‐107; Roflumilast M2‐110; Roflumilast M2‐111; Roflumilast M2‐112; Roflumilast M2‐118; Roflumilast M2‐119; Roflumilast M2‐121; Roflumilast M2‐124; Roflumilast M2‐125; Roflumilast M2‐127; Roflumilast M2‐128)
Participants See individual studies
Interventions Roflumilast 500 µg once daily
Roflumilast 250 µg once daily
Placebo once daily
Outcomes  
Notes  
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Randomised
Allocation concealment (selection bias) Unclear risk See individual studies
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trials were double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Unclear risk See individual studies
Selective reporting (reporting bias) Unclear risk See individual studies
Other bias Unclear risk See individual studies

Kavitha 2018.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: not stated
Participants Setting: 1 pulmonary medicine ward in India
Participants: 61 (intervention: 31; control: 30)
Baseline characteristics: mean age 58 years, mean FEV₁ predicted 0.93, current smokers 33%
Inclusion criteria: Indian ethnicity, age ≥ 40 years with moderate to severe COPD, current or past smokers, other co‐existing conditions
Exclusion criteria: bronchial asthma, other lung diseases, lower respiratory tract infection, pregnant or breastfeeding
Total numbers of participant withdrawals: not stated; assumed 1 person was not included in the analysis
Interventions Run‐in: not stated
  • Roflumilast 500 µg once daily with 12 µg formoterol and 9 µg tiotropium combination metered‐dose inhaler once daily

  • Formoterol 12 µg and 9 µg tiotropium combination metered‐dose inhaler once daily


Concomitant medication
  • All study participants were taking formoterol 12 µg and 9 µg tiotropium combination metered‐dose inhaler

Outcomes Primary outcomes: lung function (FEV₁ and FVC); change in mean FEV₁ after treatment
Secondary outcomes: adverse events in the roflumilast treatment group
Notes Funding not stated
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) High risk Reported as a "randomised control study". No further information about randomisation method. Also, groups are not balanced with regards to baseline characteristics. For example, the placebo group includes a high percentage of patients with diabetes
Allocation concealment (selection bias) Unclear risk Allocation concealment method was not reported
Blinding of participants and personnel (performance bias)
All outcomes High risk Assumed that there was no blinding of participants or personnel
Blinding of outcome assessment (detection bias)
All outcomes High risk Assumed that there was no blinding of the outcome assessor
Incomplete outcome data (attrition bias)
All outcomes Low risk Only 1 participant in the roflumilast group was lost; however, reasons for attrition were not reported
Selective reporting (reporting bias) High risk Outcomes not reported in the methods, so unclear whether outcomes reported were what they intended to assess. Adverse event data were not reported for the control group, so it is unclear whether there were no events in this group. Outcome data for FEV₁ were not clear, as no units were reported. If it is assumed that trial authors reported litres, then those in the intervention group improved by 660 mL, which is large in COPD terms, as it indicates 28% improvement, yet in the discussion, trial authors mention that it is similar to the 60 mL reported in the Fabbri study
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

Liu 2018.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: single hospital in Yan'an, China
Participants: 120 (roflumilast 500 µg: 60, placebo: 60)
Baseline characteristics: COPD stage II to IV according to GOLD criteria, mean age 65 years, FEV₁ % predicted < 50%, 72% male, smoking history 37 pack‐years, 66% current smokers
Inclusion criteria: aged ≥ 40 years, post‐bronchodilator FEV₁ < 50% predicted, FEV1:FVC ratio 70%, post‐bronchodilator FEV₁ with 30% to 80% predicted, COPD history > 12 months, no medication change for past 3 months
Exclusion criteria: asthma, other lung disease, systemic glucocorticosteroids, SABA 1 month before study, severe mental disorder, pregnant or breastfeeding
Total numbers of participant withdrawals: 5 (8%) and 6 (10%) from treatment and control groups, respectively
Interventions Run‐in: not stated
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Anticholingeric: 40% of participants were on anticholinergics

  • β₂‐agonist: 34% of participants were on β₂‐agonists

  • Corticosteroid: 18% of participants were on ICSs

  • LABA: not stated


23% of participants were on home oxygen therapy
Outcomes Primary outcome: change from baseline in lung function (FEV₁, FVC, and FEF25%-75%)
Secondary outcomes: quality of life (SGRQ); adverse events
Notes Funding not stated
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Randomisation was achieved via a computerised number programme generated by a statistician who was blinded to treatment allocation
Allocation concealment (selection bias) Unclear risk Unclear how the allocation sequence was concealed from patients (i.e. no mention of concealed envelopes or any other method)
Blinding of participants and personnel (performance bias)
All outcomes Low risk Participants were reported to be blinded to treatment allocation; investigators, data analysts, and outcome assessors were blinded to treatment allocation
Blinding of outcome assessment (detection bias)
All outcomes Low risk Participants were reported to be blinded to treatment allocation; investigators, data analysts, and outcome assessors were blinded to treatment allocation
Incomplete outcome data (attrition bias)
All outcomes Low risk Overall, the number of withdrawals was 9%; however there was 1 more withdrawal in the placebo group compared to the intervention group
Selective reporting (reporting bias) High risk Reporting of outcome data for lung function and SGRQ is unclear. Data provided consist of mean difference and 95% CI when standard errors should be provided. A check of the data revealed discrepancies in the numbers. Data for adverse events were also unclear. There was no reference to a protocol, so we do not know whether outcomes were reported as planned. The paper includes some confusing statements about follow‐up at 12 weeks vs 12 months ‐ probably 12 months ‐ but then follow‐up for another 3 months. Abstract states that adverse events were increased, but this is not the same as the data in the paper
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

NCT00874497 (EMPHASIS).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, triple‐blind, placebo‐controlled trial
Trial duration: 104 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 12 specialist centres across the USA
Participants: 84 (tetomilast 25 µg: 51, placebo: 33)
Baseline characteristics: mean age 58 years, 51% male
Inclusion criteria: aged 40 to 75 years, rating ≥ 1 on Goddart scale for emphysema, FEV₁:FVC > 70% predicted, ≥ 1 COPD exacerbation in the past 12 months
Exclusion criteria: asthma, active tuberculosis/bronchiectasis, respiratory tract infection in past month before screening, cancer in past 5 years, cardiovascular/endocrine blood/nervious system disorder, uncontrolled COPD exacerbation (level 2 or 3), recent systemic ICS or immunosuppressant, anticoagulant
Total numbers of participant withdrawals: 28 (54%) and 18 (54%) from treatment and control groups, respectively
Interventions Run‐in: 25 µg dose tetomilast for 2 weeks
Intervention: 50 µg once daily
Comparator: placebo once daily
Concomitant medication
  • Short‐acting anticholingeric: not stated

  • SABA: not stated

  • Corticosteroid: not stated

  • Long‐acting beta₂‐agonist: not stated

Outcomes Primary outcomes: change in FEV₁; rate of change in 20th percentile of lung voxels
Secondary outcomes: change in trough FEV₁; density mask score based on lung density voxels; rate of change in 20th percentile of lung density voxels expressed in HU units for whole lung; rate of change in emphysema (observed); change in cumulative frequency of HU; change in computed tomography (derived lung volumes); change in trough RV/TLC; change in trough inspiratory capacity; change in trough functional residual capacity; change in carbon monoxide diffusion capacity; changes in mean specific airway resistance and specific conductance; change in 7‐day average total symptom score for dyspnoea, cough, and sputum; change in 7‐day mean number of actuations of rescue medications; percentage of participants with COPD exacerbations by group; percentage of participants experiencing a COPD exacerbation
Safety outcomes: adverse events; changes in laboratory parameters, blood pressure, heart rate, physical examination findings, body weight, and BMI
Notes Clinicaltrials.gov identifier: NCT00874497
Funded by Otsuka Pharmaceutical Development & Commercialization, Inc.
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk The trial was double‐blind; participants, care providers, and outcome assessors were blinded
Blinding of outcome assessment (detection bias)
All outcomes Low risk The trial was double‐blind; participants, care providers, and outcome assessors were blinded
Incomplete outcome data (attrition bias)
All outcomes High risk 54% in each group did not complete treatment and discontinued the trial early, but there were 5 more adverse events in the tetomilast group vs the placebo group. 4 more people in the intervention arm discontinued because of early termination of the trial. Other factors included loss to follow‐up, withdrawal by either participant, or physician decision. Different numbers were used in different analyses
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Trial information was reported on the GSK website
Other bias Unclear risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

RO‐2455‐301‐RD (ACROSS).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 43 centres in mainland China, Hong Kong, and Singapore
Participants: 626 (500 µg roflumilast: 313, placebo: 313)
Baseline characteristics: mean age 64 years, 91% male, mean FEV₁ % predicted 36%, mean smoking history 37.2 pack‐years for roflumilast and 37.5 pack‐years for placebo or current smokers (24% and 29%, respectively)
Inclusion criteria: Chinese, Malaysian, or Indian ethnicity, age 40 to 80 years with severe or very severe COPD, FEV₁/FVC ≤ 0.7, post‐bronchodilator FEV₁ ≤ 50%. Current smokers or ex‐smokers with smoking history > 10 pack‐years or current smokers; 12‐month history of COPD and ≥ 14 puffs of rescue medication during the week before randomisation
Exclusion criteria: primary bronchiectasis, cystic fibrosis, bronchiolitis, lung resection, lung cancer, interstitial lung disease, active TB, lower respiratory tract infection, diagnosis of asthma at < 40 years of age, α₁‐antitrypsin deficiency
Total numbers of participant withdrawals: 67 (21.4%) and 50 (16%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind. Placebo tablets to assess suitability
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
Participants were allowed to continue taking fixed combinations of ICS plus LABA or LAMA monotherapy (e.g. tiotropium) if taken at a stable dose for at least 6 months before the run‐in period. SAMAs (e.g. ipratropium) were allowed at a constant daily dose as concomitant medication if taken on a regular basis for at least 4 weeks before study inclusion. All other COPD treatments were not allowed
Outcomes Primary outcomes: lung function; change in pre‐bronchodilator FEV₁
Secondary outcomes: changes in post‐bronchodilator FEV₁, FVC, incidence rates of COPD exacerbations, time to first COPD exacerbation, transition dyspnoea index, proportions of participants experiencing a COPD exacerbation, adverse events, changes in body weight, laboratory values, vital signs, and physical examination findings
Notes Clinicaltrials.gov identifier: NCT01313494
Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Investigators used an automated, interactive voice‐response system to randomly assign participants. The sponsor generated a list of participant numbers using a pseudo‐random number generator
Allocation concealment (selection bias) Low risk The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded, and tablets were identical in appearance
Blinding of outcome assessment (detection bias)
All outcomes Low risk The trial was double‐blinded. The investigator or anyone at the study site was prevented from knowing the treatment allocation
Incomplete outcome data (attrition bias)
All outcomes Low risk Total numbers of participants that discontinued 50 (16%) placebo, 67 (21.4%) roflumilast
Selective reporting (reporting bias) Unclear risk Outcomes were reported as planned, and the trial was registered at the NCT website
Other bias Low risk LAMA: 17.9% for placebo; 20.4% for roflumilast
SAMA: 18.2% for placebo; 17.3% for roflumilast
ICS/LABA: 55.9% for placebo; 59.7% for roflumilast
No information available. SABA allowed

RO‐2455‐402‐RD (ROBERT).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 16 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 18 centres in Denmark, Germany, Poland, Sweden, and United Kingdom
Participants: 158 (500 µg roflumilast: 79; placebo: 79)
Baseline characteristics: mean age 63 years, 77% male, mean FEV₁ predicted 60%, mean smoking history longer than 20 years or current smokers 54%
Inclusion criteria: post‐bronchodilator 30% ≤ FEV₁ ≤ 8% predicted, post‐bronchodilator FEV₁/FVC ratio ≤ 70%, current/former smoker history ≥ 20 pack‐years; aged 40 to 80 years with COPD diagnosed at least 12 months before study inclusion, chronic productive cough for 3 months in each of previous 2 years
Exclusion criteria: recent COPD exacerbation, ongoing upper or lower respiratory tract infection, asthma (with or without other lung disease), alpha‐1‐antitrypsin deficiency, bleeding disorder, concomitant glucocorticosteroids, theophylline, lipoxygenase inhibitors, antiplatelet therapy, leukotriene antagonists
Total numbers of participant withdrawals: 3 (4%) and 6 (8%) from treatment and control groups, respectively
Interventions Run‐in: 6 weeks, single‐blind with placebo to assess compliance. ICS and other non‐allowed drugs stopped
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: none

  • SABA: none

  • Corticosteroid: not permitted.

  • Long‐acting β₂‐agonist: 61% in the roflumilast group and 61% in the placebo group, respectively

Outcomes Primary outcome: change in numbers of CD8 inflammatory cells in bronchial biopsy samples
Secondary outcomes: change in numbers of inflammatory cells measured in submucosa, bronchial epithelium, induced sputum; blood FEV₁, FVC, and FEV₁/FVC ratio
Safety outcomes: adverse events; changes in laboratory parameters, blood pressure, heart rate, physical examination findings, body weight, and BMI
Notes Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk A computerised central randomisation system stratified by concomitant use of LABA was used
Allocation concealment (selection bias) Low risk Both roflumilast and placebo were given as identical yellow, triangular tablets; blinding was maintained via an interactive voice‐response system and an interactive web‐response system
Blinding of participants and personnel (performance bias)
All outcomes Low risk Participants and investigators were blinded to group assignment
Blinding of outcome assessment (detection bias)
All outcomes Low risk Outcome assessors were blinded
Incomplete outcome data (attrition bias)
All outcomes Low risk Overall the total % of participants who discontinued was 5.7%. In the roflumilast group, 3.7% discontinued compared to 7.59% in the placebo group. The number of adverse events was the same in each group; the numbers not completing were 6 in the placebo group and 3 in the roflumilast group
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. Some of the outcome data were reported in the NCT and EU clinical trials registers
Other bias Low risk No information on baseline anticholinergic, beta₂‐agonist, or corticosteroid use

RO‐2455‐404‐RD (REACT).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 203 centres in 21 countries (see online appendix)
Participants: 1945 (500 µg roflumilast: 969; placebo: 966)
Baseline characteristics: mean age 65 years, 75% male, mean FEV₁ predicted 35%, mean smoking history 48 pack‐years for roflumilast and 48 pack‐years for placebo or current smokers (42% and 45%, respectively)
Inclusion criteria: ≥ 40 years of age with a smoking history ≥ 20 pack‐years and a diagnosis of COPD with severe airflow limitation (confirmed by post‐bronchodilator FEV₁/FVC ratio < 0.70 and post‐bronchodilator FEV₁ ≤ 50% predicted), symptoms of chronic bronchitis, history of ≥ 2 exacerbations in the previous year. Participants must have been taking an ICS–LABA combination for 12 months before the study and a constant dose of an ICS–LABA fixed combination for at least 3 months before enrolment, with placebo tablet compliance of 80% to 125% during the 4‐week baseline observation period, and with a total cough and sputum score ≥ 14 (in which the score was a sum of daily scores on 4‐point scales for cough and sputum) recorded in a daily diary during the week preceding the randomisation visit
Exclusion criteria: COPD exacerbation that was ongoing during the baseline period, diagnosis of asthma or other major lung disease
Total numbers of participant withdrawals: 269 (28%) and 192 (20%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks, single‐blind. Placebo tablets to assess suitability
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
All participants used a fixed‐dose ICS–LABA combination during baseline and treatment periods. If a participant had an exacerbation that needed additional treatment during the study, the investigator could give up to 40 mg prednisolone, administered systemically, per day for 7 to 14 days. In the case of purulent sputum or suspected bacterial infection, additional antibiotic therapy was allowed. Use of the following treatments was not allowed: oral and parenteral glucocorticosteroids (except to treat acute exacerbations), LABA or ICS monotherapy, SAMA, and any SABA (with the exception of salbutamol) or oral β₂‐agonists. Participants already taking inhaled tiotropium bromide (a LAMA) were allowed to continue this treatment
Outcomes Primary outcomes: rate of moderate or severe COPD exacerbations per patient per year
Secondary outcomes: change from baseline in post‐bronchodilator FEV₁, rate of severe COPD exacerbations per patient per year, rate of COPD exacerbations per patient per year (all categories), percentage of participants experiencing ≥ 1 COPD exacerbation, time to first COPD exacerbation (all categories), time to second moderate or severe COPD exacerbation, time to third moderate or severe COPD exacerbation, number of participants needed to treat to avoid 1 moderate or severe COPD exacerbation derived from exacerbation per patient per year, number of moderate or severe COPD exacerbation days, duration of moderate or severe COPD exacerbations per participant, change from baseline in post‐bronchodilator FVC, change from baseline in post‐bronchodilator FEF (25% to 75% vital capacity), change from baseline in post‐bronchodilator FEV₆, change from baseline in post‐bronchodilator FEV₁/FVC, change from baseline in rescue medication use, change from baseline in COPD symptom score, percentage symptom‐free days, percentage rescue medication‐free days, change from baseline in CAT total score, percentage participants with CAT score improvement, time to mortality (all‐cause and COPD exacerbation‐related), time to withdrawal (all‐cause and COPD exacerbation‐related), percentage of participants with major adverse cardiovascular event, time to first major adverse cardiovascular event, percentage of participants with hospitalisation (all‐cause), time to first hospitalisation, time to withdrawal due to adverse event, percentage of participants experiencing ≥ 1 adverse event (treatment‐related), change from baseline in body weight, change from baseline in BMI
Notes Clinicaltrials.gov identifier: NCT01329029
Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Enrolled participants were randomly assigned in a 1:1 ratio, with a block size of 4, by a computerised central randomisation system, the Interactive Voice Response System–Interactive Web Response System (PPD Global Limited, Cambridge, UK)
Allocation concealment (selection bias) Low risk All parties involved in the study were masked to treatment assignment
Blinding of participants and personnel (performance bias)
All outcomes Low risk Roflumilast and placebo were supplied as identical yellow triangular tablets in wallet cards containing 40 tablets; all parties involved in the study were masked to treatment assignment
Blinding of outcome assessment (detection bias)
All outcomes Low risk All parties involved in the study were masked to treatment assignment
Incomplete outcome data (attrition bias)
All outcomes Low risk 269 participants (28%) in the roflumilast group discontinued from the study and 192 (20%) from the placebo group discontinued
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. The trial was registered at the NCT website
Other bias Unclear risk LAMA: 69% for placebo; 70% for roflumilast. No group differences stated; however 1900 (98%) of 1935 participants were using a combination of ICS–LABA according to the protocol

Roflumilast DAL‐MD‐01.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: stated
Participants Setting: single centre in USA
Participants: 27 (500 µg roflumilast: 11, placebo: 16)
Baseline characteristics: mean age 62 years, 64% male, mean FEV₁ % predicted 45%, mean smoking history 44 pack‐years for roflumilast and 47 pack‐years for placebo or current smokers (63% and 55%, respectively)
Inclusion criteria: > 40 years old with a diagnosis of moderate to severe COPD as defined by GOLD criteria, current or former cigarette smokers with more than 10 pack‐years of total consumption, chronic bronchitis defined by chronic cough and sputum production lasting ≥ 3 months for 2 consecutive years
Exclusion criteria: asthma as defined by ATS/ERS guidelines, clinically significant bronchiectasis, known sensitivity to roflumilast, use of other methylxanthines (specifically theophylline) within 1 month of screening, changes to maintenance COPD therapy within 1 month of screening
Total numbers of participant withdrawals: 1 (9%) and 1 (6%) from treatment and control groups, respectively
Interventions Run‐in: no run‐in
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
Allowed, except for theophylline. For roflumilast and placebo groups, respectively: LAMA was used by 8 (50%) and 6 (55%); ICS or LABA/ICS was used by 10 (63%) and 6 (55%)
Outcomes Primary outcome: change in induced sputum AcPGP at 12 weeks post randomisation in an intention‐to‐treat analysis
Secondary outcomes: changes in plasma AcPGP, sputum neutrophil counts, additional sputum biomarkers, 6MWT, Breathlessness Cough and Sputum Scale, SGRQ scores, changes in post‐bronchodilator FEV₁ at 12‐week visit
Notes Clinicaltrials.gov identifier NCT01572948. Funded by Forest Laboratories Inc., University of Alabama at Birmingham
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk The trial was reported as block randomised with a 1:1 allocation, stratified by current smoking status and ICS use, but no information about the sequence generation was provided
Allocation concealment (selection bias) Low risk Sealed envelopes were used to conceal allocation
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was triple‐blinded (participant, care provider, and investigator)
Blinding of outcome assessment (detection bias)
All outcomes Low risk The trial was triple‐blinded (participant, care provider, and investigator)
Incomplete outcome data (attrition bias)
All outcomes Low risk At follow‐up, only 1 participant in each group was lost because of refusal to attend the final visit or inability to be contacted for the final visit
Selective reporting (reporting bias) Low risk Outomes were reported as planned; the trial was registered at clinicaltrials.gov
Other bias Unclear risk None

Roflumilast FK1 101.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 26 weeks
Intention‐to‐treat analysis: stated
Participants Setting: not stated
Participants: 516 (roflumilast 250 µg: 175, roflumilast 500 µg: 169, placebo: 172)
Baseline characteristics: median age 61 to 62 years, 72% male, mean 38 to 63 pack‐years, 53% current smokers
Inclusion criteria: aged 40 to 75 years, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, reversibility < 12% or 200 mL, post‐bronchodilator FEV₁ 35% to 75% predicted
Exclusion criteria: not stated
Total numbers of participant withdrawals: not stated
Interventions Run‐in: 2 weeks with placebo
  • Roflumilast 500 µg once daily

  • Roflumilast 250 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: allowed at a constant daily dose for those treated before with anticholinergics on a constant dosage

  • SABA: salbutamol was allowed as rescue medication

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: post‐bronchodilator FEV₁ and FEF between 25% and 75% of vital capacity
Secondary outcomes: numbers of moderate or severe COPD exacerbations that required treatment with OCS
Notes Funding not reported
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Reported as randomised. No further information
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk Reported as double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No available information
Incomplete outcome data (attrition bias)
All outcomes Unclear risk No available information
Selective reporting (reporting bias) Unclear risk Unpublished study; no available information
Other bias Low risk None

Roflumilast FK1 103.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: not stated
Participants: 518 (roflumilast 500 µg: 200, placebo: 186)
Baseline characteristics: mean age 60 years, 75% male, 62% current smokers, average 35 pack‐years
Inclusion criteria: aged 40 to 75 years, FEV₁/FVC ≤ 0.7, post‐bronchodilator FEV₁ 35% to 75% predicted, FEV₁ reversibility ≤ 12% and ≤ 200 mL, pre‐bronchodilator FEV₁/FVC ≤ 70%
Exclusion criteria: not stated
Total numbers of participant withdrawals; not stated
Interventions Run‐in: 2 weeks with placebo
  • Roflumilast 500 µg once daily for 24 weeks

  • Roflumilast 500 µg once daily for 12 weeks. Placebo once daily for following 12 weeks


Concomitant medication
  • Short‐acting anticholinergic: all medications were withdrawn except constant‐dose short‐acting anticholinergics

  • SABA: as rescue medication

  • Corticosteroid: none

  • LABA: none


Used alongside short‐acting β₂‐agonists (available to all)
Outcomes Primary outcomes: results for 12/24‐week post‐bronchodilator FEV₁
Notes Funding not reported
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Method of randomisation not stated
Allocation concealment (selection bias) Unclear risk No information available
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No information available
Incomplete outcome data (attrition bias)
All outcomes Unclear risk No information available
Selective reporting (reporting bias) Unclear risk No information available
Other bias Low risk None

Roflumilast FLUI‐2011‐77.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 26 months
Intention‐to‐treat analysis: stated
Responder analysis for the most part
Participants Setting: 2 centres
Participants: 41 (500 µg roflumilast: 30, placebo: 11)
Baseline characteristics: not stated
Inclusion criteria: not stated
Exclusion criteria: not stated
Total numbers of participant withdrawals: not stated
Interventions Run‐in: not stated
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication: not stated
Outcomes Primary outcomes: post bronchodilation: spirometry, body plethysmography, 6MWT, patient‐reported outcomes
Secondary outcomes: not stated
Notes Clinicaltrials.gov identifier NCT01480661
Funded by Takeda
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Reported as block randomised in a 3:1 ratio of roflumilast to placebo, respectively; no further information about sequence generation
Allocation concealment (selection bias) Unclear risk No information available
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was reported as triple‐blind (participant, care provider, and investigator)
Blinding of outcome assessment (detection bias)
All outcomes Low risk The trial was reported as triple‐blind (participant, care provider, and investigator)
Incomplete outcome data (attrition bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Selective reporting (reporting bias) Low risk Outcomes reported as intended; trial registered at clinicaltrials.gov
Other bias Low risk None

Roflumilast IN‐108.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: not stated
Participants Setting: 5 centres in India
Participants: 118 recruited (roflumilast 500 µg: 47, roflumilast 200 µg: 46, placebo: 25)
Baseline characteristics: mean age 60 years, 98% male, 41% current smokers, post‐bronchodilator FEV₁ 57% to 61%, average 25 pack‐years
Inclusion criteria: not stated
Exclusion criteria: not stated
Total numbers of participant withdrawals: roflumilast 500 µg: 13 (28%); roflumilast 200 µg: 7 (15%); control 10 (40%)
Interventions Run‐in: none
  • Roflumilast 250 µg once daily

  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: not stated

  • SABA: not stated

  • Corticosteroid: none

  • LABA: not stated

Outcomes Primary outcome: post‐bronchodilator FEV₁
Secondary outcomes: COPD exacerbations, adverse events
Notes Funded by Forest Laboratories
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Reported as randomised. No further information available
Allocation concealment (selection bias) Unclear risk No information available
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was reported as double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Data as above
Selective reporting (reporting bias) Unclear risk No information available
Other bias Unclear risk None

Roflumilast JP‐706.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: not stated
Participants Setting: Japan
Participants: 600 (roflumilast 250 µg: 205, roflumilast 500 µg: 204, placebo: 191)
Baseline characteristics: mean age 70 years, 96% male, post‐bronchodilator FEV₁ not stated, average 56 pack‐years, 37% current smokers
Inclusion criteria: not stated
Exclusion criteria: not stated
Total numbers of participant withdrawals: not stated
Interventions Run‐in: single‐blind 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Roflumilast 250 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: used at a constant daily dose

  • SABA: not stated

  • Corticosteroid: not stated

  • LABA: not stated

Outcomes Primary outcomes: pulmonary function (FEV₁ pre‐bronchodilator, FVC pre‐ and post‐bronchodilator, MMEF pre‐ and post‐bronchodilator)
Secondary outcomes: number of COPD exacerbations, number of days to first COPD exacerbation, adverse events (all‐cause and drug‐related), serious adverse events (all‐cause and drug‐related)
Notes Funded by Mitsubishi‐Tanabe
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Reported as randomised; no further information about randomisation process
Allocation concealment (selection bias) Unclear risk No information available
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was reported as double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Unclear risk Not described
Selective reporting (reporting bias) High risk No further information available about the trial. No trial registry information found
Other bias Low risk None

Roflumilast M2‐107.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 159 centres in Australia, Austria, Belgium, Canada, France, Germany, Hungary, Ireland, South Africa, Spain, and the UK
Participants: 1411 (roflumilast 250 µg: 576, roflumilast 500 µg: 555, placebo: 280)
Baseline characteristics: median age 64 years, 74% male, post‐bronchodilator FEV₁ 51% for both groups, average 42 pack‐years, 45% current smokers
Inclusion criteria: aged ≥ 40 with history of COPD > 12 months, FEV₁ < 50% predicted, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, reversibility < 12% or 200 mL, mean post‐bronchodilator FEV₁ 30% to 80% predicted
Exclusion criteria: asthma, lung cancer or bronchiectasis, long‐term oxygen treatment, recent exacerbation that required a course of systemic corticosteroids, emergency room treatment or hospital admission within 4 weeks before run‐in period
Total numbers of participant withdrawals: 124 (22%) and 32 (11%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Roflumilast 250 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: used at a constant daily dose

  • SABA: salbutamol as rescue medication

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: post‐bronchodilator FEV₁; SGRQ total score
Secondary outcomes: change from baseline in pre‐bronchodilator FEV₁; post‐bronchodilator FVC; post‐bronchodilator FEV in 6 seconds and FVC; FEF rate between 25% and 75% of vital capacity; number of moderate or severe COPD exacerbations
Notes Funded by ALTANA Pharma AG
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk The randomisation sequence was generated by the sponsor in a blind manner
Allocation concealment (selection bias) Unclear risk No information on allocation concealment method, but "no person involved in data analysis had knowledge of the randomisation sequence"
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind. Roflumilast and placebo tablets and packaging were identical, so neither participants nor study personnel were aware of either medication allocation
Blinding of outcome assessment (detection bias)
All outcomes Low risk "No person involved in data analysis had knowledge of the randomisation sequence"
Roflumilast and placebo tablets and packaging were identical, so the investigator was not aware of either medication allocation
Incomplete outcome data (attrition bias)
All outcomes Low risk 100 participants discontinued from the roflumilast 250 µg group, 124 from the roflumilast 500 µg group, and 32 from the placebo group
Selective reporting (reporting bias) High risk There was inconsistency in the quoting of statistical errors. Within the text and in Table 2, data are quoted as "least squares means and SD"; however in Figures 2 and 3, SE bars are shown. It is more likely that results represented SE, not SD. Trial registration was not found
Other bias Low risk None

Roflumilast M2‐110.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: not stated
Participants Setting: 36 centres in Argentina, Canada, Columbia, Mexico, Peru, and the USA
Participants: 909 participants randomised (roflumilast 500 µg: 449; placebo: 460)
Baseline characteristics: aged between 55 and 74 years (mean age 64.2 years in the roflumilast group and 64.6 years in the placebo group), 88% participants were white, roflumilast group included 51% males and the placebo group 55% males
Inclusion criteria: clinical diagnosis of COPD based on ATS criteria, post‐bronchodilator FEV₁/FVC ≤ 70%, post‐bronchodilator FEV₁ ≥ 30% and ≤ 80% predicted, post‐bronchodilator FEV₁ increase ≤ 12% or ≤ 200 mL compared to pre‐bronchodilator value, score grade ≥ 1 on the MRC Dyspnea Scale, currently stable COPD with no change in COPD treatment in the prior 4 weeks
Exclusion criteria: clinical diagnosis of asthma, poorly controlled COPD, regular need for daily oxygen therapy
Total numbers of participant withdrawals: roflumilast group: 15.4% withdrew due to adverse events, 10.5% withdrew consent, 2.9% withdrew due to lack of efficacy; placebo group: 7.6% withdrew due to adverse events, 8.5% withdrew consent, 3% withdrew due to protocol violation
Interventions Run‐in: 4‐week single‐blind period during which respiratory medication (including ICS, LABA, and long‐acting anticholinergics) was withdrawn
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: none

  • SABA: none

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: pulmonary function tests (FEV₁, FVC, FEF, PEF, FIV₁, FVCin)
Secondary outcomes: exacerbation rate; quality of life; symptoms; use of rescue medication; safety and tolerability
Notes ClinicalTrials.gov Identifier: NCT00062582. Funded by ALTANA Pharma AG
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Reported as randomised. Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Low risk Assumed that the allocation concealment method was adequate due to pharmaceutical company sponsorship
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was reported as double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Unclear risk Roflumilast group: 15.4% withdrew due to adverse events, 10.5% withdrew consent, 2.9% withdrew due to lack of efficacy; placebo group: 7.6% withdrew due to adverse events, 8.5% withdrew consent, 3% withdrew due to protocol violation
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. The trial was registered at clinicaltrials.gov

Roflumilast M2‐111.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: M2‐111 was conducted at 188 centres in 6 countries, and M2‐112 at 159 centres in 14 countries
Participants: 1176 participants were randomised in this study (roflumilast: 500 µg: 568; placebo: 608)
Baseline characteristics: severe COPD according to GOLD criteria grades III and IV, mean age 64 to 65 years, 72% male
Inclusion criteria: aged ≥ 40 years, post‐bronchodilator FEV₁ < 50% predicted, reversibility < 15%, mean post‐bronchodilator FEV₁ 42%, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, 40% current smokers, 60% ex‐smokers, average 46 to 48 pack‐years
Exclusion criteria: history of asthma, lung cancer, or bronchiectasis; need for long‐term oxygen therapy; known α₁‐antitrypsin deficiency, clinically significant cardiopulmonary comorbidity
Total numbers of participant withdrawals: data combined with M2‐112 showing 433 (33%) and 348 (26%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: 891 patients on short‐acting anticholinergics

  • SABA: salbutamol as rescue medication

  • Corticosteroid: 943 patients continued corticosteroid use

  • LABA: none


Used alongside corticosteroids, anticholinergics, and rescue short‐acting β₂‐agonists 54% overall (available to all)
Outcomes Primary outcomes: change from baseline to endpoint in post‐bronchodilator FEV₁; number of moderate or severe exacerbations per patient per year
Secondary outcomes: change from baseline in SGRQ total score; change from baseline in pre‐bronchial FEV₁, post‐bronchodilator FEV in 6 seconds and in FVC; FEF rate between 25% and 75% vital capacity; number of moderate or severe COPD exacerbations requiring systemic corticosteroid treatment per patient per year
Notes NCT00076089/BY217/M2‐111. Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk The randomisation sequence was generated by a multiplicative congruent pseudo‐random numbers generator programme (programme RANDOM, based on Fishman and Moore)
Allocation concealment (selection bias) Low risk "Each study participant who qualified was assigned a number in sequential order. Code labelling prevented the investigator and the patient from knowing which drug was administered"
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Data combined with M2‐112
Selective reporting (reporting bias) Low risk Trial registered at clinicaltrials.gov; outcomes reported as planned. M2‐111 and M2‐112 data combined
Other bias Low risk None

Roflumilast M2‐111+M2‐112.

Study characteristics
Methods As described in separate studies above and below
Participants  
Interventions  
Outcomes  
Notes  
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk See individual trials
Allocation concealment (selection bias) Low risk See individual trials
Blinding of participants and personnel (performance bias)
All outcomes Low risk See individual trials
Blinding of outcome assessment (detection bias)
All outcomes Low risk See individual trials
Incomplete outcome data (attrition bias)
All outcomes Low risk See individual trials
Selective reporting (reporting bias) Low risk See individual trials
Other bias Low risk  

Roflumilast M2‐112.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 159 centres in 14 countries
Participants: 1514 (roflumilast 500 µg: 761, placebo: 753)
Baseline characteristics: severe COPD according to GOLD criteria grades III and IV, mean age 65 years, 75% male
Inclusion criteria: aged ≥ 40 years, post‐bronchodilator FEV₁ < 50% predicted, reversibility < 15%, mean post‐bronchodilator FEV₁ 41%, FEV₁/FVC ≤ 0.7 with smoking history > 10 pack‐years, 37% current smokers, 63% ex‐smokers, average 44 pack‐years
Exclusion criteria: history of asthma, lung cancer, or bronchiectasis; need for long‐term oxygen therapy; known α₁‐antitrypsin deficiency or clinically significant cardiopulmonary comorbidity
Total numbers of participant withdrawals: 217 (29%) and 163 (22%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: 891 participants on short‐acting anticholinergics

  • SABA: salbutamol as rescue medication

  • Corticosteroid: 943 participants continued corticosteroid use

  • LABA: none


Used alongside corticosteroids, anticholinergics, and rescue short‐acting β₂‐agonists 54% overall (available to all)
Outcomes Primary outcomes: change from baseline to endpoint in post‐bronchodilator FEV₁ and in the number of moderate or severe exacerbations per patient per year
Secondary outcomes: change from baseline in SGRQ total score; change from baseline in pre‐bronchial FEV₁; post‐bronchodilator FEV in 6 seconds and FVC; FEF rate between 25% and 75% of vital capacity; number of moderate or severe COPD exacerbations requiring systemic corticosteroid treatment per patient per year
Notes NCT00430729/BY217/M2‐112. Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk "The randomisation list was generated using a multiplicative congruent pseudo‐random number generator (program RANDOM, based on Fishman and Moore)"
Allocation concealment (selection bias) Low risk "Each study participant who qualified was assigned a number in sequential order. Code labelling prevented the investigator and the patient from knowing which drug was administered"
Blinding of participants and personnel (performance bias)
All outcomes Low risk The trial was double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk "Over 70% of patients completed the study. The reasons for withdrawal were similar between groups except for adverse events, which occurred more frequently with roflumilast"
"Withdrawal due to COPD exacerbations was reported in 3.5 and 3.2% of patients in roflumilast and placebo groups, respectively"
Selective reporting (reporting bias) Low risk None
Other bias Low risk None

Roflumilast M2‐118.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 22 centres in 4 countries
Participants: 250 (roflumilast 500 µg: 127, placebo: 123)
Baseline characteristics: mean age 60 years, 73% (roflumilast) vs 84% (placebo) male, post‐bronchodilator FEV₁ 55% predicted, average 41 pack‐years, 53% current smokers
Inclusion criteria: clinically stable patients ≥ 40 years of age with smoking history > 10 pack‐years and 12‐month history of COPD. Other inclusion criteria included post‐bronchodilator FEV₁ 30% to 80% predicted, FEV₁/forced vital capacity (FVC) < 0.7, and set plethysmographic FRC and peak oxygen uptake requirements
Exclusion criteria: asthma or lung disease other than COPD, α₁‐antitrypsin deficiency, participation in pulmonary rehabilitation programme within 2 months, supplemental oxygen therapy, significant medical comorbidity that may influence exercise tolerance
Total numbers of participant withdrawals: 16 (13%) and 12 (10%) from treatment and control groups, respectively
Interventions Run‐in: 2‐ to 3‐week baseline period consisting of 2 familiarisation visits during which a symptom‐limited constant work rate cycle exercise test was performed at 75% of maximum incremental work rate. If a constant work rate endurance time was not produced within 2 minutes at both visits, a third visit was performed. If reproducibility was not achieved at the third visit, the patient was not randomised
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: participants could use ipratropium bromide in regular stable doses as needed

  • SABA: participants could use short‐acting β₂‐agonists as needed

  • Corticosteroid: ICSs were permitted throughout the study if taken at a constant dosage for > 3 months before the start of the study

  • LABA: none

Outcomes Primary outcomes: activity‐related dyspnoea (TDI); spirometry and body plethysmography; symptom‐limited exercise tests
Notes Funded by Nycomed GmbH (Konstanz, Germany)
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Reported as randomised. Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk Trial reported as double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Of 250 randomised participants, 16 from the roflumilast group and 12 from the placebo group discontinued prematurely
Selective reporting (reporting bias) Unclear risk Outcomes reported, but no trial protocol found on trial registry websites
Other bias Low risk None

Roflumilast M2‐119.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 32 centres in 5 countries
Participants: 410 (roflumilast 500 µg: 203, placebo: 207)
Baseline characteristics: mean age 68 years, 93% male, post‐bronchodilator FEV₁ 50.5% predicted, average 44 pack‐years, 69% current smokers
Inclusion criteria: former or current smokers with pack‐year history ≥ 10, aged ≥ 40 years, post‐bronchodilator FEV₁/FVC ≤ 0.7, FEV₁ 30% to 80% predicted, clinically stable COPD within 4 weeks before baseline
Exclusion criteria: history of asthma or other relevant lung disease, COPD exacerbation within 4 weeks before baseline, need for long‐term oxygen therapy, known α₁‐antitrypsin deficiency, clinically significant cardiopulmonary comorbidity
Total numbers of participant withdrawals: 40 (20%) and 18 (9%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: "short‐acting anticholinergics at a constant daily dosage as concomitant medication if already taken on a regular basis at a constant dosage for at least 4 weeks prior to the study"

  • SABA: patients could use SABAs as needed

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcome: mean change in post‐bronchodilator FEV₁ from baseline
Secondary outcomes: mean change in pre‐bronchodilator FEV₁ from baseline; change in other lung function measures, time to COPD exacerbation; proportion of participants experiencing exacerbations; time to study withdrawal; adverse effects
Notes Clinicaltrials.gov identifier: NCT00242320; BY217/M2‐119. Funded by Nycomed GmbH, Konstanz, Germany
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Computer‐generated randomisation list used
Allocation concealment (selection bias) Unclear risk Not stated
Blinding of participants and personnel (performance bias)
All outcomes Low risk Double‐blinded
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes High risk Of the 411 randomised participants, 41 from the roflumilast group and 18 from the placebo group discontinued during the treatment period (20% compared with 8%, respectively)
Selective reporting (reporting bias) Low risk Outcomes were reported as planned. The protocol was registered at clincaltrials.gov
Other bias Low risk None

Roflumilast M2‐121.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 12 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 16 centres in 6 countries
Participants: 600 participants (full analysis set; roflumilast 500 µg: 301, placebo: 299)
Baseline characteristics: median age 65 years, 74% male, FEV₁ 46% predicted, 48 mean pack‐years
Inclusion criteria: history of COPD ≥ 12 months as defined by GOLD criteria, age ≥ 40 years, FEV₁/FVC ratio (post‐bronchodilator) ≤ 70%, FEV₁ (post‐bronchodilator) ≤ 65% predicted, FRC (post‐bronchodilator) ≤ 120% predicted
Exclusion criteria: COPD exacerbation indicated by treatment with systemic glucocorticosteroids not stopped ≥ 4 weeks before baseline visit; non‐smoker, current smoker, or ex‐smoker (smoking cessation ≥ 1 year ago) with smoking history < 10 pack‐years; any concomitant disease that might interfere with study procedures or evaluation
Total numbers of participant withdrawals: 32 participants withdrew due to COPD exacerbations
Interventions Run‐in: 4‐week single‐blind placebo tablet once daily in the morning and all disallowed concomitant medications withdrawn
  • Roflumilast 500 µg once daily

  • Placebo


Concomitant medication
  • Short‐acting anticholinergic: none

  • SABA: none

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcome: lung function parameters indicative of hyperinflation in people with COPD
Secondary outcomes: mean change from randomisation to endpoint in additional pre‐ and post‐bronchodilator spirometric and lung volume parameters; measurement of quality of life parameters; dyspnoea
Notes ClinicalTrials.gov Identifier: NCT00108823; BY217/M2‐121. Funded by AstraZeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Reported as randomised. Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk Trial reported as double‐blind
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk No available information
Incomplete outcome data (attrition bias)
All outcomes Low risk Of 600 randomised participants, 13 from the roflumilast group and 19 from the placebo group withdrew due to exacerbations
Selective reporting (reporting bias) Unclear risk A publication was not found for this trial; however, study results were obtained from the trial registry website
Other bias Low risk None

Roflumilast M2‐124.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 246 centres in 10 countries
Participants: 1513 (roflumilast 500 µg: 760, placebo: 753)
Baseline characteristics: mean age 64 years, 71% male, post‐bronchodilator FEV₁ 37.6% predicted, average 47 pack‐years, 48% current smokers
Inclusion criteria: former or current smokers with ≥ 20 pack‐year history, aged ≥ 40 years, post‐bronchodilator FEV₁/FVC ≤ 0.7, chronic cough and sputum production, post‐bronchodilator FEV₁ < 50% predicted, ≥ 1 recorded COPD exacerbation requiring systemic glucocorticosteroids or treatment in hospital in previous year
Exclusion criteria: available in the online web appendix (p 11)
Total numbers of participant withdrawals: 264 (34%) and 234 (31%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: 31% of those in the roflumilast group and 32% in the placebo group

  • SABA: "patients could use short acting β₂‐agonists as needed"

  • Corticosteroid: none

  • LABA: "eligible patients were stratified according to their use of long acting β₂‐agonists and smoking status"; roflumilast 49%, placebo 51%

Outcomes Primary outcomes: mean change in pre‐bronchodilator FEV₁; mean rate of COPD exacerbations requiring oral or parenteral glucocorticosteroids or requiring hospitalisation or leading to death (per patient per year)
Secondary outcomes: mean change in post‐bronchodilator FEV₁; time to mortality for any reason; natural log‐transformed CRP (mg/L); mean TDI focal score
Notes Clinicaltrials.gov identifier: NCT00297102. Funded by AstraZeneca
Adverse event data are pooled with numbers from study M2‐125, which followed an identical study design
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk A randomisation list was generated via a pseudo‐random numbers generator, and an automated interactive voice‐response system was used to randomly assign participants
Allocation concealment (selection bias) Low risk All individuals involved in the studies were unaware of treatment assignment
Blinding of participants and personnel (performance bias)
All outcomes Low risk "All individuals involved in the studies were unaware of treatment assignment. Tablets were identical in appearance. The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk 264 participants discontinued from the roflumilast group and 234 discontinued from the placebo group
Selective reporting (reporting bias) Low risk Outcomes reported as planned. Trial registered at clinicaltrials.gov
Other bias High risk 44% of participants in both roflumilast and placebo groups received corticosteroid pre‐treatment

Roflumilast M2‐124+M2‐125.

Study characteristics
Methods As described in separate studies above and below
Participants  
Interventions  
Outcomes  
Notes  
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk See individual studies
Allocation concealment (selection bias) Low risk See individual studies
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk See individual studies
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk See individual studies
Incomplete outcome data (attrition bias)
All outcomes Unclear risk See individual studies
Selective reporting (reporting bias) Unclear risk See individual studies
Other bias Unclear risk See individual studies

Roflumilast M2‐125.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 221 centres in 8 countries
Participants: 1571 (roflumilast 500 µg: 773, placebo: 798)
Baseline characteristics: mean age 64 years, 80% male, average 48 pack‐years, 35% current smokers
Inclusion criteria: former or current smokers with pack‐year history ≥ 20 years, aged ≥ 40 years, post‐bronchodilator FEV₁/FVC ≤ 0.7, chronic cough and sputum production, post‐bronchodilator FEV₁ < 50% predicted, ≥ 1 recorded COPD exacerbation requiring systemic glucocorticosteroids or treatment in hospital in previous year
Exclusion criteria: available in the online web appendix (p 11)
Total numbers of participant withdrawals: 246 (32%) and 248 (31%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: 38% of those in the roflumilast group and 41% of the placebo group

  • SABA: "patients could use short acting β₂‐agonists as needed"

  • Corticosteroid: none

  • LABA: "eligible patients were stratified according to their use of long acting β₂‐agonists and smoking status"; roflumilast 48%, placebo 51%

Outcomes Primary outcomes: mean change in pre‐bronchodilator FEV₁; mean rate of COPD exacerbations (moderate or severe) requiring oral or parenteral glucocorticosteroids or requiring hospitalisation or leading to death (per patient per year)
Secondary outcomes: mean change in post‐bronchodilator FEV₁; time to mortality for any reason; natural log‐transformed CRP (mg/L); mean TDI focal score during treatment period
Notes Clinicaltrials.gov identifier: NCT00297115; BY217/M2‐125. Funded by AstraZeneca
Adverse event data are pooled with numbers from study M2‐124, which followed an identical study design
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk A randomisation list was generated via a pseudo‐random numbers generator, and an automated interactive voice‐response system was used to randomly assign participants
Allocation concealment (selection bias) Low risk All individuals involved in the studies were unaware of treatment assignment
Blinding of participants and personnel (performance bias)
All outcomes Unclear risk "All individuals involved in the studies were unaware of treatment assignment. Tablets were identical in appearance. The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling"
Blinding of outcome assessment (detection bias)
All outcomes Unclear risk Assumed that this would be low risk; however, no available information
Incomplete outcome data (attrition bias)
All outcomes Low risk 246 patients discontinued from the roflumilast group and 248 discontinued from the placebo group
Other bias High risk 40% of participants in both roflumilast and placebo groups received corticosteroid pre‐treatment

Roflumilast M2‐127.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 135 centres in 10 countries
Participants: 1221 (roflumilast 500 µg: 467, placebo: 468)
Baseline characteristics: mean age 65 years, 71% male, post‐bronchodilator FEV₁ 54.7% and 55.3% predicted (roflumilast and placebo), average 43 pack‐years, 39% current smokers
Inclusion criteria: former or current smokers with ≥ 1 year smoking cessation and a pack‐year history ≥ 10, aged ≥ 40 years, post‐bronchodilator FEV₁/FVC ≤ 0.7, post‐bronchodilator FEV₁ 40% to 70% predicted, partial reversibility to albuterol with increase from baseline FEV₁ ≤ 12% or 200 mL
Exclusion criteria: available in the online web appendix (p 10)
Total numbers of participant withdrawals: 107 (23%) and 82 (18%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo once a day
  • Roflumilast 500 µg and salmeterol once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: none

  • SABA: participants used short‐acting β₂ as rescue medication

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: change in mean pre‐bronchodilator FEV₁ from baseline to each post‐randomisation visit
Secondary outcomes: post‐bronchodilator FEV₁ and FVC; TDI score; SOBQ; rate of COPD exacerbations; use of rescue medication
Notes ClinicalTrials.gov identifier NCT00313209; BY217/M2‐127; 2005‐005080‐28 (EudraCT Number). Funded by Nycomed GmbH, Konstanz, Germany
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk "The sponsor generated a randomisation list of patient random numbers using a pseudo‐random number generator. The investigator used an automated, interactive voice response system to randomly assign patients"
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk All individuals involved in the studies were unaware of treatment assignment
Blinding of outcome assessment (detection bias)
All outcomes Low risk All individuals involved in the studies were unaware of treatment assignment
Incomplete outcome data (attrition bias)
All outcomes Low risk 107 participants discontinued from the roflumilast group and 82 discontinued from the placebo group
Selective reporting (reporting bias) Low risk Outcomes reported as planned. Trial registered at clincialtrials.gov
Other bias Low risk None

Roflumilast M2‐128.

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 85 centres in 7 countries
Participants: 910 (roflumilast 500 µg: 372, placebo: 372)
Baseline characteristics: mean age 64 years, 71% male, post‐bronchodilator FEV₁ 56.0% and 56.2% predicted (roflumilast and placebo), average 44 pack‐years, 40% current smokers
Inclusion criteria: former or current smokers with ≥ 1 year smoking cessation and a pack‐year history ≥ 10, aged ≥ 40 years, post‐bronchodilator FEV₁/FVC ≤ 0.7, post‐bronchodilator FEV₁ 40% to 70% predicted, partial reversibility to albuterol with increase from baseline FEV₁ ≤ 12% or 200 mL
Exclusion criteria: available in the online web appendix (p 10)
Total numbers of participant withdrawals: 62 (17%) and 39 (11%) from treatment and control groups, respectively
Interventions Run‐in: 4 weeks with placebo once a day
  • Roflumilast 500 µg and tiotropium once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholingeric: none

  • SABA: participants used short‐acting β₂ as rescue medication

  • Corticosteroid: none

  • LABA: none

Outcomes Primary outcomes: change in mean pre‐bronchodilator FEV₁ from baseline to each post‐randomisation visit
Secondary outcomes: post‐bronchodilator FEV₁ and FVC; TDI score; SOBQ; rate of COPD exacerbations; use of rescue medication
Notes Clinicaltrials.gov identifier: NCT0042468; BY217/M2‐128. Funded by Nycomed GmbH, Konstanz, Germany
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk "The sponsor generated a randomisation list of patient random numbers using a pseudo‐random number generator. The investigator used an automated, interactive voice response system to randomly assign patients"
Allocation concealment (selection bias) Low risk The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling
Blinding of participants and personnel (performance bias)
All outcomes Low risk "All individuals involved in the studies were unaware of treatment assignment. The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling. Tablets were identical in appearance"
Blinding of outcome assessment (detection bias)
All outcomes Low risk "All individuals involved in the studies were unaware of treatment assignment. The investigator or anyone at the study site was prevented from knowing the allocation sequence with code labelling. Tablets were identical in appearance"
Incomplete outcome data (attrition bias)
All outcomes Low risk 62 participants discontinued from the roflumilast group and 39 discontinued from the placebo group
Selective reporting (reporting bias) Unclear risk Outcomes reported as planned. Trial protocol registered at clincialtrials.gov and at European trial registry
Other bias Low risk None

Roflumilast ROF‐MD‐07(RE2SPOND).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, double‐blind, placebo‐controlled trial
Trial duration: 52 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 338 locations across Australia, Argentina, Canada, Chile, Columbia, Italy, Malaysia, Peru, Phillippines, Romania, Russia, Serbia, Spain, Taiwan, and Ukraine
Participants: 2354 (500 µg roflumilast: 1178; placebo: 1176)
Baseline characteristics: mean age 64 years, 68% male, mean FEV₁ % predicted 33%, mean smoking history 52.2 pack‐years for roflumilast and 53.1 pack‐years for placebo or current smokers (39% and 40%, respectively)
Inclusion criteria: ≥ 40 years with severe or very severe COPD, chronic bronchitis, ≥ 2 exacerbations and/or hospitalisations in the previous year, receiving ICS/LABA with or without LAMA daily for ≥ 3 months
Exclusion criteria: within the 4 weeks before enrolment, had a moderate or severe COPD exacerbation and/or a COPD exacerbation treated with antibiotics or systemic corticosteroids or a lower respiratory tract infection. Other exclusionary criteria included diagnoses of other lung diseases, moderate to severe liver impairment (Child‐Pugh B or C), HIV or hepatitis infection, current diagnosis of asthma, cancer in the past 5 years, α₁‐antitrypsin deficiency, clinically significant cardiovascular condition, resting QTc interval > 470 ms, BMI ≥ 45 kg/m²
Total numbers of participant withdrawals: 337 (29%) and 254 (21%) from treatment and control groups, respectively
Interventions Run‐in: 2 weeks, single‐blind. Placebo tablets to assess suitability
  • Roflumilast 500 µg once daily

  • Placebo once daily


Concomitant medication
ICS/LABA FDC (fluticasone propionate/salmeterol, 250/50 mg (1 inhalation twice a day), or budesonide/formoterol, 160/4.5 mg (2 inhalations twice a day)). Participants taking fluticasone propionate/salmeterol, 500/50 mg, at study entry were required to switch to the lower dosage (250/50 mg) before entry. Up to 60% of participants were allowed concomitant LAMA (e.g. tiotropium) if administered for ≥ 3 months before screening, with no dose change. Those not on LAMA were allowed a SAMA
Outcomes Primary outcome: rate of moderate or severe COPD exacerbations per patient per year
Secondary outcomes: rate of COPD exacerbations leading to hospitalisation or death (severe COPD exacerbations); rate of moderate or severe exacerbations; rate of moderate or severe COPD exacerbations or COPD exacerbations treated with antibiotics; rate of moderate or severe COPD exacerbations treated with antibiotics during the treatment period; mean change in pre‐dose FEV₁; mean change in pre‐dose FEV₁ from randomisation over 52 weeks; adverse events; mortality (all‐cause); serious adverse events; other adverse events (not including serious events)
Notes Clinicaltrials.gov identifier: NCT01443845. Funded by Astra Zeneca
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Low risk Reported as randomised. Assumed that the randomisation method was adequate due to pharmaceutical company sponsorship
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk Reported as triple‐blind (participant, investigator and outcome assessor)
Blinding of outcome assessment (detection bias)
All outcomes Low risk Reported as triple‐blind (participant, investigator and outcome assessor)
Incomplete outcome data (attrition bias)
All outcomes Unclear risk 337 participants (29%) discontinued from the roflumilast group and 254 (22%) discontinued from the placebo group
Selective reporting (reporting bias) Unclear risk Outcomes reported as planned. Trial registered at clinicaltrials.gov
Other bias Unclear risk 47% in each treatment group were taking LAMAs. Participants were also using combined LABA/ICS. 65% in the placebo group were taking fluticasone propionate/salmeterol FDC, and 65% in the roflumilast group. 35% of participants in each treatment group were taking budesonide/formoterol FDC

Urban 2018 (ELASTIC).

Study characteristics
Methods Study design: parallel‐group study
Randomisation: randomised, triple‐blind, placebo‐controlled trial
Trial duration: 24 weeks
Intention‐to‐treat analysis: stated
Participants Setting: 1 specialist respiratory and critical care medicine centre at a hospital in Austria
Participants: 80 (roflumilast 500 µg: 40, placebo: 40)
Baseline characteristics: median age 64, 52% male, median 50 smoking pack‐years
Inclusion criteria: 40 years of age and over, history of ≥ 2 COPD exacerbation requiring systemic corticosteroid treatment or hospitalisation in the last year
Exclusion criteria: inability to comply with study medication, history of acute exacerbation, alpha₁‐antitrypsin deficiency, asthma, acute/severe respiratory infection, lung cancer, bronchiectasis, ILD, acute MI, systolic left ventricular dysfunction, CHF, cardiac arrhythmia/heart valve deformation, peripheral arterial occlusive disease, acute or chronic hepatic failure, autoimmune disease, active malignancy, pregnant/breastfeeding, hypersensitivity to study medication or placebo, mental or neurological disorder, history of depression
Total numbers of participant withdrawals: 7 (17%) and 6 (15) in the roflumilast and placebo groups, respectively
Interventions Run‐in: 4 weeks
  • Roflumilast 500 µg, once daily

  • Placebo once daily


Concomitant medication
  • Short‐acting anticholinergic: not stated

  • SABA: not stated

  • Corticosteroid: not stated

  • LABA: not stated

Outcomes Primary outcome: change in carotid femoral‐pulse wave velocity
Secondary outcomes: change in reactive hyperaemia index; change in augmentation index; change in matrix metalloproteinase‐9; change in asymmetrical dimethylamine; change in tumour necrosis factor‐alpha; change in FEV₁; change in 6‐minute walk test; change in COPD assessment test
Notes Clincaltrials.gov identifier: NCT01630200. Funded by Ludwig Boltzmann Institute for COPD and Respiratory Epidemiology
Risk of bias
Bias Authors' judgement Support for judgement
Random sequence generation (selection bias) Unclear risk Reported as randomised; no further information
Allocation concealment (selection bias) Unclear risk No available information
Blinding of participants and personnel (performance bias)
All outcomes Low risk "All responsible persons, those administering interventions or assessing the outcomes, and elementally all experimental and control patients were blinded to group assignment"
Blinding of outcome assessment (detection bias)
All outcomes Low risk "All responsible persons, those administering interventions or assessing the outcomes, and elementally all experimental and control patients were blinded to group assignment"
Incomplete outcome data (attrition bias)
All outcomes Low risk 40 randomised in each group. In the roflumilast group 33/40 completed (82.5%), in the placebo arm 34/40 completed (85%), so similar attrition. Similar numbers of non‐fatal and serious fatal adverse events in each group
Selective reporting (reporting bias) Low risk Study authors reported outcomes as planned; methods and results were published on EU trials registry
Other bias Unclear risk Criteria for COPD not well defined apart from exacerbations

6MWT: 6‐minute walk test; AcPGP: plasma acetyl‐proline‐glycine‐proline; ATS: American Thoracic Society; BDI: Baseline Dyspnoea Index; BMI: body mass index; BORG Scale: rating of perceived exertion; CAT: COPD Assessment Test; CHF: congestive heart failure; COPD: chronic obstructive pulmonary disease; CRP: C‐reactive protein; DLCO: diffusing capacity of the lung for carbon monoxide; EU: European Union; ERS: European Respiratory Society; FDC: fixed dose combination; FEF: forced expiratory flow; FEV₁: forced expiratory volume in one second; FEV₆: forced expiratory volume in six seconds; FIV₁: forced expiratory volume in one second; FRC: functional residual capacity; FVC: forced vital capacity; GOLD: Global Initiative for Chronic Obstructive Lung Disease; HU: Hounsfield unit; HIV: human immunodeficiency virus; IC: inspiratory capacity; ICS: inhaled corticosteroid; ILD: interstitial lung disease; LABA: long‐acting beta₂‐agonist; LAMA:long‐acting muscarinic antagonist; MDI: metered‐dose inhaler; MI: myocardial infarction; MMEF: maximal mid‐expiratory flow; NCT: national clinical trial; PEF: peak expiratory flow; QTc: corrected Q wave and T wave; RFRC: resting functional residual capacity; RV: residual volume; SABA: short‐acting beta₂‐agonist; SAMA: short‐acting muscarinic antagonist; SD: standard deviation; SE: standard error; SGRQ: St George's Respiratory Questionnaire; SOBQ: Shortness of Breath Questionnaire; SVC: slow vital capacity; TDI: transition dyspnoea index; TLC: total lung capacity VO₂: oxygen uptake.

Characteristics of excluded studies [ordered by study ID]

Study Reason for exclusion
Borker 2003 Insufficient data; only RR of QoL improvement provided
CTRI/2012/09/002961 No placebo group
CTRI/2014/01/004370 No placebo group
Ferguson 2003 Integrated results from four 24‐week cilomilast trials
Fischer 2003 Analysis focused on participants with baseline SGRQ score ≥ median SGRQ score only
Grootendorst 2001 Endpoint: first dose bronchodilator effects only
Grootendorst 2002 Treatment Bayer BAY 19‐8004; 11 participants; only 1 week in duration
Grootendorst 2003 Endpoint: first dose bronchodilator effects only
Grootendorst 2007 Cross‐over design
GSK256066 Phase 2 trial; no primary outcome measure investigating lung function; only 1 trial to date
Kelsen 2002 No study ID or group numbers identified
Knobil 2003 No SD or SE given
Lim 2004 Combining results from 2 pivotal European phase 3 cilomilast trials
NCT00246935 Different regimens of roflumilast; no placebo group
NCT01849341 Different regimens of roflumilast; no placebo group
NCT01973998 Patients were diagnosed with AECOPD
NCT02018432 Different regimens of roflumilast; no placebo group
Nieman 1999 Study 038: insufficient data available for changes in lung function and exacerbation rates
Pascoe 2007 Treatment QAK423 (Novartis), discontinued. Only 1 trial available
Rabe 2017 Editorial letter
Reisner 2003 Combined results; individual studies already included in review
Rennard 2008 Systematic review; relevant individual studies already included in review
Roflumilast JP708 JP108 is an extension study of APTA‐2217‐06. After the key open of APTA‐2217‐06, administration to placebo group would be terminated. Not all participants enrolled in JP106 continued onto the JP108 study
Sadigov 2014 No placebo group
Sadigov 2015 Open‐label; no placebo group
SB207499/040 Open‐label study; men or women with COPD who successfully completed study 042 or 091 in which participants received cilomilast 15 mg twice daily or placebo for 24 weeks in study 042 and 26 weeks in study 091 without tolerability problems. Concomitant COPD medication use allowed; given placebo or placebo/Ariflo during study period
SB207499/041 Open‐label study; men or women with COPD who successfully completed study 039 in which participants received cilomilast 15 mg twice daily or placebo for 24 weeks without tolerability problems. Concomitant COPD medication use allowed; given placebo or placebo/Ariflo during study period
Song 2005 Abstract only; unable to contact study author
Spencer 2002 No study identification or group numbers identified
Vestbo 2007 Treatment UK‐500,001 (Pfizer); discontinued
Vestbo 2009 Treatment UK‐500,001 (Pfizer); discontinued
Wang 2005 Although quoted as significant, mean and SD figures not provided
Watz 2013 Inhaled therapy
Watz 2016 Different regimens of roflumilast

AECOPD: acute exacerbation of COPD; COPD: chronic obstructive pulmonary disease; QoL: quality of life; RR: risk ratio; SD: standard deviation; SE: standard error; SGRQ: St George's Respiratory Questionnaire.

Characteristics of studies awaiting classification [ordered by study ID]

Barnes 2014.

Methods international, 16‐week, randomised, double‐blind, placebo‐controlled, parallel‐group study investigating effects of roflumilast 500 µg once daily vs placebo on inflammatory parameters in bronchial biopsy tissue specimens, sputum, and blood serum
Participants 150 participants with COPD and chronic bronchitis for at least 12 months will be recruited into the study and randomised in a 1:1 ratio to receive either roflumilast or placebo
Interventions Roflumilast and placebo
Outcomes Primary endpoint will be number of CD8+ cells in bronchial biopsy tissue specimens (submucosa)
Key secondary endpoint will be number of CD68+ cells assessed by indirect immunohistochemistry
Notes Completed; awaiting results

EUCTR2004‐004442‐40‐GB.

Methods Randomised controlled trial
Participants Participants with history of moderate to severe COPD for at least 12 months
Interventions Roflumilast and placebo
Outcomes Primary outcome variable will be mean change in post‐bronchodilator FEV₁ from baseline
Notes No data provided; awaiting results

Mahmud 2013.

Methods Single‐blind, randomised, placebo‐controlled study carried out in the Department of Respiratory Medicine at National Institute of Diseases of the Chest and Hospital (NIDCH), Dhaka, Bangladesh
Participants 130 participants were recruited initially and were randomly distributed into Group A, where they received conventional therapy (inhaled salmeterol + fluticasone and tiotropium) and roflumilast (0.5 mg once daily), and Group B, where participants were given placebo with conventional therapy
Study duration was 3 months
Interventions As above
Outcomes Primary outcome variable was change in mean FEV₁
Secondary outcome variable was change in mean CAT score from baseline
Notes No data provided; study authors contacted

NCT00671073.

Methods Multi‐centre double‐blind randomised controlled trial over 12 weeks across USA investigating the safety and efficacy of various doses of oglemilast
Participants 427 participants with COPD, post‐bronchodilator FEV₁/FVC < 70%, post‐bronchodilator FEV₁ > 30% and < 80%
Interventions Oglemilast and placebo
Outcomes Primary outcome variable will be pre‐bronchodilator morning (trough) FEV₁ at 12 weeks
Secondary endpoint will be pre‐bronchodilator FVC at 12 weeks
Notes No data provided; awaiting results

NCT01595750.

Methods Single‐centre, double‐blind randomised controlled trial over 12 weeks in Spain
Participants 150 participants with a diagnosis of COPD, FEV₁ < 70%; current and former smokers
Interventions Roflumilast and placebo
Outcomes Primary outcome variable will be endothelial function at 12 weeks
Secondary endpoints include arterial stiffness; serum and plasma inflammation markers; serum oxidative stress markers; serum endothelial dysfunction markers at 12 weeks
Notes No data provided; awaiting results

NCT01701934.

Methods Triple‐blind randomised controlled trial for 26 weeks investigating whether roflumilast could improve metabolic profiles and reduce visceral adiposity in people with COPD
Participants 14 participants with moderate to severe COPD, assigned to either 500 µg roflumilast or placebo for 26 weeks
Interventions Roflumilast and placebo
Outcomes Primary outcome variables will be change in body mass index; change in waist circumference; change in hip‐to‐waist ratio; change in metabolic profiles; change in body composition; change in subcutaneous adiposity; change in liver fat
Notes No data provided; awaiting results

CAT: COPD Assessment Test; COPD: chronic obstructive pulmonary disease; FEV₁: forced expiratory volume in one second; FVC: forced vital capacity.

Characteristics of ongoing studies [ordered by study ID]

NCT02451540 2015.

Study name Evaluation of the effect of roflumilast in hyperinflated COPD patients using functional respiratory imaging
Methods Parallel RCT
Participants 40 people who are stable on LABA/LAMA therapy and who are prone to dynamics hyperinflation
Interventions Roflumilast and placebo
Outcomes Radiological (CT) changes in airway measures
Changes in spirometry and body plethysmography
Starting date September 2015
Contact information University Hospital of Antwerp
Notes Other Study ID Numbers: FLUI‐2014‐134, EudraCT
Estimated study completion date: January 2017

NCT02671942 2016.

Study name A multicenter randomised double‐blind clinical study evaluated the safety, pharmacokinetic and pharmacodynamic characteristics of roflumilast in COPD patients
Methods Parallel RCT
Participants People with COPD in China
Interventions Roflumilast and placebo
Outcomes Area under the plasma concentration after vs drug dose
Percentage of participants with adverse events of interest
Change in pre‐bronchodilator FEV₁ during the down‐titration period
Starting date March 2016
Contact information Contact: Zheng Jinping
Notes Estimated enrolment: 120
Estimated study completion date: August 2017

COPD: chronic obstructive pulmonary disease; CT: computed tomography; FEV₁: forced expiratory volume in one second; LABA: long‐acting beta₂‐agonist; LAMA:long‐acting muscarinic antagonist; RCT: randomised controlled trial.

Differences between protocol and review

We added the comparison between published and unpublished results when we discovered the large number of unpublished studies but before we extracted data from the studies and carried out the analysis.

We excluded cross‐over trials, as carry‐over effects and disease progression cannot be adequately controlled for in people with COPD.

We updated the methods section in accordance with MECIR standards.

We separated mortality from non‐fatal serious adverse events in the methods section for clarity of presentation in the 'Summary of findings' table and in other sections of the review.

Contributions of authors

Phillippa Poole: protocol initiation and development, checking of content of current update, corresponding author.

Sadia Janjua: screening, data extraction, risk of bias assessment, and write‐up of the 2020 review update.

Rebecca Fortescue: data extraction and risk of bias assessment.

Contributions of editorial team

Chris Cates (Co‐ordinating Editor): checked data entry before the full write‐up of the review, edited the protocol, advised on methods, and approved the updated review prior to publication.

Emma Dennett (Managing Editor): co‐ordinated the editorial process, advised on interpretation and content, and edited the review.

Emma Jackson (Assistant Managing Editor): conducted peer review and edited various sections and references in the protocol and in the review.

Elizabeth Stovold (Information Specialist): designed the search strategy, ran the searches, and edited the search methods section.

Sources of support

Internal sources

  • University of Auckland provided salary support for Professor Phillippa Poole, New Zealand

External sources

  • The authors declare that no such funding was received for this systematic review, Other

Declarations of interest

Phillippa Poole: none known.

Sadia Janjua is funded full‐time as a systematic reviewer by a National Institute for Health Research (NIHR) Programme Grant to complete work on this review.

Rebecca Fortescue is Co‐ordinating Editor for Cochrane Airways.

New search for studies and content updated (no change to conclusions)

References

References to studies included in this review

Cilomilast 039 {published data only}

  1. 207499/039. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of cilomilast (15 mg twice daily) in patients with chronic obstructive pulmonary disease (207499/039). gsk-clinicalstudyregister.com/study/207499/039#rs (first received 28 September 2008).
  2. Edelson JD, Compton C, Nieman R, Robinson CB, Amit O, Bagchi I, et al. Cilomilast (Ariflo), a potent selective phosphodiesterase 4 inhibitor, reduces exacerbations in COPD patients: results of a 6 month trial. American Journal of Respiratory and Critical Care Medicine 2001;163(5 Suppl):A771. [Google Scholar]
  3. Edelson JD, Compton C, Nieman R, Robinson CB, Watt R, Amit O, et al. Cilomilast (Ariflo) improves health status in patients with COPD: results of a 6-month trial. American Journal of Respiratory and Critical Care Medicine 2001;163(5 Suppl):A277. [Google Scholar]
  4. Rennard SI, Schachter N, Strek M, Rickard K, Amit O. Cilomilast for COPD: results of a 6-month, placebo controlled study of a potent, selective inhibitor of phosphodiesterase 4. Chest 2006;129(1):55-66. [DOI] [PubMed] [Google Scholar]

Cilomilast 042 {unpublished data only}

  1. 207499/042. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of cilomilast (15 mg twice daily) in patients with chronic obstructive pulmonary disease. gsk-clinicalstudyregister.com/study/207499/042?search=study&search_terms=cilomilast&search=Search#rs (first received 28 September 2008).

Cilomilast 076 {published and unpublished data}

  1. 207499/076. A 12-week, multicentre, double-blind, placebo-controlled, parallel-group study to evaluate the anti-inflammatory activity of SB207499 15 mg twice daily in patients with chronic obstructive pulmonary disease. gsk-clinicalstudyregister.com/files/pdf/24047.pdf (first received 28 September 2008).
  2. Gamble E, Grootendorst DC, Brightling CE, Troy S, Qiu Y, Zhu J, et al. Antiinflammatory effects of the phosphodiesterase-4 inhibitor cilomilast (Ariflo) in chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2003;168:976-82. [DOI] [PubMed] [Google Scholar]

Cilomilast 091 {unpublished data only}

  1. 207499/091. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study followed by a 2-week, randomized, double-blind, run-out phase to evaluate the efficacy, safety, tolerability and discontinuation of SB207499 (15 mg twice daily) in patients with chronic obstructive pulmonary disease. gsk-clinicalstudyregister.com/study/207499/091?search=study&search_terms=SB207499#rs (first received 28 September 2008).

Cilomilast 103657 {unpublished data only}

  1. CIL103657. GSK CTR-657. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of cilomilast (15 mg BID) in patients with chronic obstructive pulmonary disease (COPD). gsk-studyregister.com/study?uniqueStudyId=CIL103657 (first received 24 August 2016).

Cilomilast 110 {unpublished data only}

  1. 207499/110. A 12-week, multicenter, double-blind, placebo-controlled, parallel-group study to evaluate the anti-inflammatory activity of cilomilast 15 mg twice daily in patients with chronic obstructive pulmonary disease. gsk-studyregister.com/study/5979 (first received 11 December 2008).

Cilomilast 111 {published and unpublished data}

  1. 207499/111. A 12-week, randomized, double-blind, placebo-controlled, parallel-group study to investigate the effect of cilomilast (15 mg twice daily) on trapped gas volume in patients with chronic obstructive pulmonary disease. gsk-clinicalstudyregister.com/files/pdf/24050.pdf (first received 28 September 2008).
  2. Zamel N, McClean P, Zhu J, Schryver B, Madan A, Robinson CB, et al. Effect of cilomilast (Ariflo) on trapped gas volume and indices of hyperinflation in patients with chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2002;165(Suppl 8):A226. [Google Scholar]

Cilomilast 121 {unpublished data only}

  1. SB207499/121. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of cilomilast (15 mg BID) in patients with chronic obstructive pulmonary disease. h/s3.amazonaws.com/ctr-gsk-7381/207499_121/430f519b-3d76-4244-9417-3bc801497511/ef158d16-ce04-46dd-8358-456dfee5641f/24042-v1.pdf (first received 28 September 2008).

Cilomilast 156 {unpublished data only}

  1. 207499/156. A randomized, 24-week, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of cilomilast (15 mg BID) in patients with chronic obstructive pulmonary disease. s3.amazonaws.com/ctr-gsk-7381/207499_156/7dd49801-f278-4112-bbe8-f8ef3c62ce78/f7a0f0ba-f28a-4666-b76a-f155eec89363/gsk-207499-156-clinical-study-report-redact-v1.pdf (first received 20 May 2015).

Cilomilast 157 {unpublished data only}

  1. 207499/157. A randomised, double-blind, placebo-controlled, parallel-group study to evaluate the efficacy, safety and tolerability of oral cilomilast (15 mg bd) when given as maintenance treatment for 12 months to subjects with chronic obstructive pulmonary disease. s3.amazonaws.com/ctr-gsk-7381/207499_157/37e6fc79-d3ca-46e1-b07a-c7c7995d5c69/31ef48f1-c164-406b-b9db-cf88a0d606dc/gsk-207499-157-clinical-study-report-redact-v1.pdf (first received 28 September 2008).

Cilomilast 168 {published and unpublished data}

  1. 207499/168. A randomized, 12-week, double-blind, placebo-controlled, parallel-group study to evaluate the safety and tolerability of cilomilast 15 mg twice daily in patients with chronic obstructive pulmonary disease. gsk-clinicalstudyregister.com/files/pdf/24054.pdf (first received 28 September 2008).
  2. Reisner C, Zhu J, Morris A, Lim J, Knobil K. Assessment of cardiac events via 24-hour electrocardiographic (Holter) monitoring with cilomilast in chronic obstructive pulmonary disease. European Respiratory Journal 2003;22(Suppl 45):P522. [Google Scholar]
  3. Reisner C, Zhu J, Morris A, Lim J, Knobil K. Assessment of cardiac events via 24-hour electrocardiographic (Holter) monitoring with cilomilast in chronic obstructive pulmonary disease. In: American Thoracic Society 99th International Conference; 2003 May 16-21; Seattle. 2003.

Cilomilast 180 {unpublished data only}

  1. 207499/180. An 18-week randomized, double-blind, placebo-controlled, multicenter study designed to compare treatment with cilomilast to that with placebo for changes in ventilatory mechanics and function (both at rest and during exercise), as well as related exertional dyspnea and exercise performance, in hyperinflated patients with stable COPD. s3.amazonaws.com/ctr-gsk-7381/207499_180/07613483-8e07-4a60-9f7c-fc673665e770/b6f75a99-2ac4-4a74-9b06-b5f74ec8c056/gsk-207499-180-clinical-study-report-redact-v1.pdf (first received 20 November 2008).

Cilomilast 181 {unpublished data only}

  1. 207499/181. A 13-week randomised, double-blind, parallel group, multicentre study to compare the bronchial anti-inflammatory activity of oral cilomilast (15 mg bd) with placebo twice daily in subjects with chronic obstructive pulmonary disease. s3.amazonaws.com/ctr-gsk-7381/207499_181/ac461889-1b68-44f0-a3ef-e3f7a9c76c93/cdcf6d43-b6ec-4d91-a018-d183ceb8b804/gsk-207499-181-clinical-study-report-redact-v1.pdf (first received 28 September 2008).

Compton 2001 {published and unpublished data}

  1. Compton CH, Gubb J, Cedar E, Bakst A, Nieman RB, Amit O, et al. SB 207499, a second generation, oral PDE₄ inhibitor, improves health status in patients with COPD. In: European Respiratory Society Annual Congress; 1999 Oct 9-13; Madrid. 1999:P2237.
  2. Compton CH, Gubb J, Nieman R, Edelson J, Amit O, Bakst A, et al. Cilomilast, a selective phosphodiesterase-4 inhibitor for treatment of patients with chronic obstructive pulmonary disease: a randomised, dose-ranging study. Lancet 2001;358(9278):265-70. [DOI] [PubMed] [Google Scholar]

COPD safety pool {published data only}

  1. Durmowicz AG. Cross discipline team leader review, application number 022522Orig1s000. www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022522Orig1s000CrossR.pdf (accessed prior to 14 February 2020).

Kavitha 2018 {published data only}

  1. Kavitha DM, Sarumathy S, Sasidharan SL, Shaik M, Sandeep G, Rajasekhar V, et al. A clinical study on safety and efficacy of formoterol and tiotropium combination compared to formoterol and tiotropium with roflumilast combination in treatment of moderate to severe chronic obstructive pulmonary disease patients. Asian Journal of Pharmaceutical and Clinical Research 2018;11(3):184-6. [Google Scholar]

Liu 2018 {published data only}

  1. Liu MBD-Y, Wang MMZ-G, YG MB, Zhang MMH-M, Zhang MMY-X, Wang MBX-J, et al. Effect and safety of roflumilast for chronic obstructive pulmonary disease in Chinese patients. Medicine 2018;97(9):e9864. [DOI] [PMC free article] [PubMed] [Google Scholar]

NCT00874497 (EMPHASIS) {published data only}

  1. NCT00874497. Pilot study of tetomilast in chronic obstructive pulmonary disease (COPD) associated with emphysema (EMPHASIS) [A multicentre, randomised, double-blind, placebo-controlled pilot study to assess the pharmacodynamics, efficacy and safety of 50mg tetomilast administered as oral tablets in patients with chronic obstructive pulmonary disease associated with emphysema]. clinicaltrials.gov/ct2/show/NCT00874497 (first received 2 April 2009).

RO‐2455‐301‐RD (ACROSS) {published data only}

  1. NCT01313494. A chronic obstructive pulmonary disease (COPD) trial investigating roflumilast on safety and effectiveness in China, Hong Kong and Singapore. https://clinicaltrials.gov/show/NCT01313494 2011.
  2. Zheng J, Yang J, Zhou X, Zhao L, Hui F, Wang H, et al. Roflumilast for the treatment of COPD in an Asian population: a randomized, double-blind, parallel-group study. Chest 2014;145(1):44-52. [CENTRAL: 978808] [EMBASE: 2014049205] [4900126000007427] [PMID: ] [DOI] [PubMed] [Google Scholar]

RO‐2455‐402‐RD (ROBERT) {published data only}

  1. 2011-000582-13. A 16-week, randomised, placebo-controlled, double-blind, and parallel group trial to assess the anti-inflammatory effects of roflumilast in chronic obstructive pulmonary disease. The ROBERT study. clinicaltrialsregister.eu/ctr-search/trial/2011-000582-13/results (first received 4 January 2012).
  2. NCT01509677. Trial to assess the anti-inflammatory effects of roflumilast in chronic obstructive pulmonary disease. https://clinicaltrials.gov/show/NCT01509677 .
  3. Rabe KF, Hanauer G, Strigun A, Alagappan V. Effect of roflumilast on the serum metabolome of COPD patients. American Journal of Respiratory and Critical Care Medicine 2018;197:A7429. [Google Scholar]
  4. Rabe KF, Henrik W, Baraldo S, Pedersen F, Biondini D, Bagul N. Anti-inflammatory effects of roflumilast in chronic obstructive pulmonary disease (ROBERT): a 16-week, randomised, placebo-controlled trial. Lancet Respiratory Medicine 2018;6:827-36. [DOI] [PubMed] [Google Scholar]
  5. Rabe KF, Saetta M, Watz H, Baraldo S, Hanauer G, Göhring U-M, et al. Reduction in airway eosinophils in patients with COPD treated with roflumilast for 16 weeks: a double-blind, parallel-group, randomised, placebo-controlled biopsy trial. American Journal for Respiratory and Critical Care Medicine 2017;195:A7569. [Google Scholar]

RO‐2455‐404‐RD (REACT) {published data only}

  1. Calverley PM, Rabe KF, Goehring U, Kristiansen S, Kristiansen S, Martinez FJ. Does roflumilast decrease exacerbations in severe COPD patients not controlled by inhaled combination therapy? The REACT study protocol. International Journal of COPD 2012;7(1):375-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Kiff C, Ruiz S, Varol N, Gibson D, Davies A, Purkayastha D. Cost-effectiveness of roflumilast as add-on to triple inhaled therapy versus triple inhaled therapy in patients with severe and very severe chronic obstructive pulmonary disease associated with chronic bronchitis in the UK. International Journal of Chronic Obstructive Pulmonary Disease 2018;13(5):2707-20. [DOI: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Martinez FJ, Calverley PMA, Goehring U-M, Brose M, Fabbri LM, Rabe KF. Effect of roflumilast on exacerbations in patients with severe chronic obstructive pulmonary disease uncontrolled by combination therapy (REACT): a multicentre randomised controlled trial. Lancet 2015;385(9971):857-66. [DOI] [PubMed] [Google Scholar]
  4. NCT01329029. Effect of roflumilast on exacerbation rate in patients with COPD treated with fixed combinations of LABA and ICS. A 52-week, randomised double-blind trial with roflumilast 500 µg versus placebo. The REACT trial. clinicaltrials.gov/show/NCT01329029 (first received 30 March 2011).

Roflumilast DAL‐MD‐01 {published data only}

  1. Wells JM, Jackson PL, Viera L, Bhatt SP, Gautney J, Handley G, et al. A randomized, placebo-controlled trial of roflumilast. Effect on proline-glycine-proline and neutrophilic inflammation in chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2015;192(8):934-42. [CENTRAL: 1077156] [EMBASE: 2015481225] [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Wells JM, Viera L, Gautney J, Handley GH, Jackson PL, Bhatt SP, et al. A randomized, placebo-controlled trial of roflumilast on markers of inflammation in chronic obstructive pulmonary disease (COPD). American Journal of Respiratory and Critical Care Medicine 2015;191(Meeting Abstracts):A3643. [EMBASE: 72051470] [DOI] [PMC free article] [PubMed] [Google Scholar]

Roflumilast FK1 101 {published and unpublished data}

  1. Bredenbroker D, Syed J, Leichtl S, Rathgeb F, Wurst W. Roflumilast, a new, orally active phosphodiesterase 4 inhibitor, is effective in the treatment of chronic obstructive pulmonary disease. In: European Respiratory Society Annual Congress; 2002 14-18 Sep; Stockholm. 2002.
  2. Bredenbroker D, Syed J, Leichtl S, Rathgeb F, Wurst W. Safety of once-daily roflumilast, a new, orally active, selective phosphodiesterase 4 inhibitor, in patients with COPD. American Journal of Respiratory and Critical Care Medicine 2002;165(Suppl 8):A595. [Google Scholar]
  3. Leichtl S, Syed J, Bredenbröker D, Rathgeb F, Wurst W. Efficacy of once-daily roflumilast, a new, orally active, selective phosphodiesterase 4 inhibitor, in chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2002;165(Suppl 8):A229. [Google Scholar]
  4. Leichtl S, Syed J, Bredenbroker D, Rathgeb F, Wurst W. Roflumilast, a new, orally active, selective phosphodiesterase 4 inhibitor, is safe and well tolerated in patients with chronic obstructive pulmonary disease. In: European Respiratory Society Annual Congress; 2002 Sep 14-17; Stockholm. 2002.

Roflumilast FK1 103 {published and unpublished data}

  1. Boszormenyi-Nagy G, Pieters WR, Steffen H, Timar M, Vinkler I, Teichmann P, et al. The effect of roflumilast treatment and subsequent withdrawal in patients with COPD. In: American Thoracic Society International Conference; 2005 May 20-25; San Diego. 2005.
  2. Rabe K, Similowski T, Bredenbröker D, Teichmann P, Böszörményi-Nagy G. Onset of action and effect of withdrawal of roflumilast in COPD. In: European Respiratory Society Annual Congress; 2011 Sep 24-28; Amsterdam. 2011.

Roflumilast FLUI‐2011‐77 {published data only}

  1. De Backer J, Vos W, Claes R, Hufkens A, Bedert L, De Backer W. A double blind placebo controlled study to assess the effect of roflumilast in addition to LABA/LAMA/ICS treatment in COPD patients using novel biomarkers. American Journal of Respiratory and Critical Care Medicine 2014;189:A3773. [CENTRAL: 1035550] [Google Scholar]
  2. De Backer J, Vos W, Van Holsbeke C, Claes R, Hufkens A, Verplancke V, et al. A double blind placebo controlled study to assess the effect of roflumilast in addition to LABA/LAMA/ICS treatment in COPD patients using novel biomarkers. American Journal of Respiratory and Critical Care Medicine 2014;44(Suppl 58):4670. [CENTRAL: 1053499] [EMBASE: 72043284] [Google Scholar]
  3. De Backer W, Vos W, Van Holsbeke C, Vinchurkar S, Claes R, Hufkens A, et al. The effect of roflumilast in addition to LABA/LAMA/ICS treatment in COPD patients. European Respiratory Journal 2014;44(2):527-9. [CENTRAL: 998328] [EMBASE: 2014530222] [PMID: ] [DOI] [PubMed] [Google Scholar]

Roflumilast IN‐108 {unpublished data only}

  1. Brown P. Clinical pharmacology and biopharmaceutics review(s). Application number 022522Orig1s000. accessdata.fda.gov/drugsatfda_docs/nda/2011/022522Orig1s000ClinPharmR.pdf (accessed 9 October 2019).

Roflumilast JP‐706 {unpublished data only}

  1. Brown P. Clinical pharmacology and biopharmaceutics review(s). Application number 022522Orig1s000. www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022522Orig1s000ClinPharmR.pdf (accessed 9 October 2019).

Roflumilast M2‐107 {published and unpublished data}

  1. Bateman ED, Holmes M, Muir JF, Andrae K, Witte S, Bredenbroeker D. Safety profile of roflumilast, a novel, selective phosphodiesterase 4 inhibitor, in patients with moderate to severe COPD. In: American Thoracic Society 100th International Conference; 2004 May 21-26; Orlando. 2004.
  2. O'Donnell D, Muir JF, Jenkins C, Plit P, Brockhaus F, Witte S, et al. Roflumilast, a novel selective phosphodiesterase 4 inhibitor, improves quality of life and lowers exacerbation rate in patients with moderate to severe COPD [Abstract]. In: American Thoracic Society 100th International Conference; 2004 May 21-26; Orlando. 2004 Orlando.
  3. Rabe F, O'Donnell D, Muir F, Jenkins C, Witte S, Bredenbroeker D, et al. Roflumilast an oral once daily PDE4 inhibitor improves lung function and reduces exacerbation rates in patients with COPD. European Respiratory Journal 2004;24(Suppl 48):21s. [Google Scholar]
  4. Rabe KF, Bateman ED, O'Donnell D, Witte S, Bredenbröker D, Bethke TD. Roflumilast - an oral anti-inflammatory treatment for chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 2005;36(9485):563-71. [DOI] [PubMed] [Google Scholar]
  5. Rabe KF, Chapman KR, Joubert J, Vetter N, Witte S, Bredenboecker D. Roflumilast, a novel, selective phosphodiesterase 4 inhibitor, improves lung function in patients with moderate to severe COPD. In: American Thoracic Society 100th International Conference; 2004 May 21-26; Orlando. 2004.
  6. Rabe KF, O'Donnell D, Bateman ED, Andrae K, Witte S, Bredenbroeker D. Roflumilast improves lung function and quality of life in chronic obstructive pulmonary disease. Chest 2004;126(4 Suppl):709S-a. [Google Scholar]

Roflumilast M2‐110 {unpublished data only}

  1. NCT00062582. Effect of roflumilast on pulmonary function and respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD) (BY217/M2-110) [A 24 week, placebo-controlled, randomized, parallel group study comparing roflumilast 500 mcg daily vs placebo on pulmonary function and respiratory symptoms in patients with chronic obstructive pulmonary disease (COPD)]. clinicaltrials.gov/ct2/show/study/NCT00062582 (accessed prior to 23 June 2017).

Roflumilast M2‐111 {published data only}

  1. NCT00076089. OPUS study: effect of roflumilast on exacerbation rate in patients with chronic obstructive pulmonary disease (BY217/M2-111). https://clinicaltrials.gov/show/NCT00076089 .
  2. Rennard SI, Calverley PM, Goehring UM, Bredenbroker D, Martinez FJ. Reduction of exacerbations by the PDE4 inhibitor roflumilast - the importance of defining different subsets of patients with COPD. Respiratory Research 2011;12:18. [1465-993X: (Electronic). 1465-9921 (Linking)] [http://filehosting.pharmacm.com/DownloadService.ashx?client=CTR_MED_7111&studyid=4477&filename=BY217-M2-111-RDS-2008-12-23.pdf ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Rennard SI, Calverley PMA, Rempel A, Bredenbroker D, Martinez FJ. The effect of roflumilast treatment on exacerbations in patients with COPD results of a pooled analysis of two 1-year studies. In: American Thoracic Society International Conference; 2008 May 16-21; Toronto. 2008.
  4. Rusch H, Gooss A, Bethke TD, Rennard S. Efficacy of roflumilast when used with concomitant inhaled corticosteroids from the OPUS/RATIO studies. Respiration 2011;82(1):67-107. [Google Scholar]

Roflumilast M2‐111+M2‐112 {published data only}

  1. Rennard SI, Calverley PM, Goehring UM, Bredenbroker D, Martinez FJ. Reduction of exacerbations by the PDE4 inhibitor roflumilast - the importance of defining different subsets of patients with COPD. Respiratory Research 2011;12:18. [1465-993X: (Electronic). 1465-9921 (Linking)] [DOI] [PMC free article] [PubMed]
  2. Rennard SI, Calverley PMA, Rempel A, Bredenbroker D, Martinez FJ. The effect of roflumilast treatment on exacerbations in patients with COPD results of a pooled analysis of two 1-year studies. In: American Thoracic Society International Conference; 2008 May 16-21; Toronto. 2008.
  3. Rusch H, Gooss A, Bethke TD, Rennard S. Efficacy of roflumilast when used with concomitant inhaled corticosteroids from the OPUS/RATIO studies. Respiration 2011;82(1):67-107. [Google Scholar]

Roflumilast M2‐112 {published and unpublished data}

  1. Calverley PM, Fabbri LM, Teichmann P, Bredenbroeker D. Effect of roflumilast on lung function and exacerbations in patients with chronic obstructive pulmonary disease: results of a one year study. Thorax 2005;2(Suppl II):ii42. [Google Scholar]
  2. Calverley PM, Sanchez-Toril F, McIvor A, Teichmann P, Bredenbroeker D, Fabbri LM. Effect of 1-year treatment with roflumilast in severe chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2007;176(2):154-61. [DOI] [PubMed] [Google Scholar]
  3. Calverley PM, Sanchez-Toril F, McIvor RA, Teichmann P, Bredenbroeker D, Fabbri LM. Effect of roflumilast on lung function: a 1-year study in patients with severe to very severe COPD. In: Proceedings of the American Thoracic Society; 2006 May 19-24; San Diego. 2006.
  4. Fabbri LM, Sanchez-Toril F, McIvor RA, Teichmann P, Bredenbroeker D, Calverley PM. Effect of roflumilast on exacerbations: a 1-year study in patients with severe to very severe COPD. In: American Thoracic Society Conference; 2006 May 19-24; San Diego. 2006.
  5. Mclvor RA, Calverley PM, Sanchez-Toril F, Teichmann P, Bredenbroeker D, Fabbri LM. Effect of roflumilast on quality of life: a 1-year study in patients with severe to very severe COPD. In: American Thoracic Society Conference; 2006 May 19-24; San Diego. Vol. 3. 2006:A850.
  6. Rennard SI, Calverley PM, Goehring UM, Bredenbroker D, Martinez FJ. Reduction of exacerbations by the PDE4 inhibitor roflumilast - the importance of defining different subsets of patients with COPD. Respiratory Research 2011;12:18. [1465-993X: (Electronic). 1465-9921 (Linking)] [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Rusch H, Gooss A, Bethke TD, Rennard S. Efficacy of roflumilast when used with concomitant inhaled corticosteroids from the OPUS/RATIO studies. Respiration 2011;82(1):67-107. [Google Scholar]
  8. Rutten-van Molken M, Van Nooten F, Lindermann M, Caser M. The 1-year cost effectiveness of roflumilast for the treatment of severe to very severe COPD patients. European Respiratory Journal 2007;30(Suppl 51):194s, P1188. [Google Scholar]
  9. Rutten-van Molken MP, Nooten FE, Lindemann M, Caeser M, Calverley PM. A 1-year prospective cost-effectiveness analysis of roflumilast for the treatment of patients with severe chronic obstructive pulmonary disease. PharmacoEconomics 2007;25(8):695-711. [CENTRAL: 610748] [PMID: ] [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐118 {published data only}

  1. O'Donnell DE, Bredenbroker D, Brose M, Webb KA. Physiological effects of roflumilast at rest and during exercise in COPD. European Respiratory Journal 2012;39(5):1104-12. [ES:1399-3003: IL:0903-1936] [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐119 {published data only}

  1. Hui D, Mahayiddin A, Roa C, Kwa KH, Bredenbröker D, Goehring UM, et al. Roflumilast in Asian patients with COPD: a randomised placebo-controlled trial. In: European Respiratory Society Annual Congress; 2011 Sep 24-28; Amsterdam. 2011. [DOI] [PubMed]
  2. Lee JS, Hong YK, Park TS, Lee SW, Oh Y-M, Lee S-D. Efficacy and safety of roflumilast in Korean patients with COPD. Yonsei Medical Journal 2016;57(4):928-35. [CENTRAL: 1158901] [EMBASE: 20160381439] [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Lee SD, Hui DS, Mahayiddin AA, Roa CC, Kwa KH, Goehring UM, et al. Roflumilast in Asian patients with COPD: a randomized placebo-controlled trial. Respirology 2011;16(8):1249-57. [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐121 {unpublished data only}

  1. NCT00108823. The HERO-study: effects of roflumilast in patients with COPD (Chronic Obstructive Pulmonary Disease) (BY217/M2-121) [A 24-week, double blind, randomized study to investigate the effect of 500 µg roflumilast tablets once daily versus placebo on parameters indicative of hyperinflation in patients with chronic obstructive pulmonary disease]. clinicaltrials.gov/ct2/show/NCT00108823 (first received 19 April 2005).

Roflumilast M2‐124 {published and unpublished data}

  1. Calverley PM, Rabe KF, Goehring U-M, Kristiansen S, Fabbri LM, Martinez FJ, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009;374(9691):685-94. [DOI] [PubMed] [Google Scholar]
  2. Martinez F, Hanania N, AURA Study Team. Efficacy and safety of the phosphodiesterase-4 inhibitor roflumilast in patients with symptomatic chronic obstructive pulmonary disease in the M2-124 study. Chest 2009;136(4):3S-e. [Google Scholar]
  3. NCT00297102. Effect of roflumilast on exacerbation rate in patients with chronic obstructive pulmonary disease (COPD): the AURA study (BY217/M2-124). https://clinicaltrials.gov/show/NCT00297102 .
  4. Nowak D, Ehlken B, Kotchie R, Wecht S, Magnussen H. Roflumilast in combination with long-acting bronchodilators. Deutsche Medizinische Wochenschrift 2013;138(4):119-25. [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐124+M2‐125 {published data only}

  1. Bateman ED, Rabe KF, Calverley PMA, Goehring UM, Brosee M, Bredenbroker D, et al. Roflumilast with long-acting beta2-agonists for COPD: influence of exacerbation history. European Respiratory Journal 2011;38(3):553-60. [DOI] [PubMed] [Google Scholar]
  2. Calverley P, Fabbri L, Rabe K, Goehring UM, Martinez F. Efficacy of the PDE4 inhibitor roflumilast in COPD patients with chronic bronchitis. In: European Respiratory Society Annual Congress; 2009 Sep 12-16; Vienna. 2009.
  3. Calverley P, Martinez F, Goehring UM, Bredenbröker D, Brose M, Vogelmeier C. Impact of roflumilast treatment on the rate and duration of exacerbations and overall steroid load in patients with COPD. In: European Respiratory Society Annual Congress; 2011 Sep 24-28; Amsterdam. 2011.
  4. Calverley PM, Rabe KF, Goehring U-M, Kristiansen S, Fabbri LM, Martinez FJ, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009;374(9691):685-94. [DOI] [PubMed] [Google Scholar]
  5. Calverley PMA, Rabe KF, Goehring UM, Kristiansen S, Fabbri LM, Martinez FJ. Erratum: Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials (The Lancet (2009) 374 (685-694)). Lancet 2010;376(9747):1146. [DOI] [PubMed] [Google Scholar]
  6. Gooss A, Rusch H, Bethke TD, Hanania N. Efficacy of roflumilast in patients receiving concomitant treatments for chronic obstructive pulmonary disease over 12 months. Respiration 2011;82(1):67-107. [Google Scholar]
  7. Hanania NA, Brose M, Larsson T, Rabe KF. Efficacy of roflumilast in patients receiving concomitant treatments for chronic obstructive pulmonary disease over 12 months. American Journal of Respiratory and Critical Care Medicine 2010;181:A4435. [Google Scholar]
  8. Hanania NA, Calverley PMA, Dransfield MT, Karpel JP, Brose M, Zhu H, et al. Pooled subpopulation analyses of the effects of roflumilast on exacerbations and lung function in COPD. Respiratory Medicine 2014;108(2):366-75. [CENTRAL: 985699] [EMBASE: 2014100136] [PMID: ] [DOI] [PubMed] [Google Scholar]
  9. Kaplan A, Calverley P. Efficacy of roflumilast in patients with symptomatic chronic obstructive pulmonary disease (COPD) receiving concomitant bronchodilator treatments. Primary Care Respiratory Journal 2010;19(2):A13 [50]. [Google Scholar]
  10. Martinez F, Fabbri L, Rabe K, Goehring U-M, Calverley P. Safety of the PDE4 inhibitor roflumilast in COPD patients with chronic bronchitis [Abstract]. In: European Respiratory Society Annual Congress; 2009 Sep 12-16; Vienna. 2009.
  11. Martinez FJ, Rabe KF, Goehring UM, Lakkis H, Rowe P, Palm U. Roflumilast prolongs time to first and subsequent exacerbations in patients with severe to very severe COPD. American Journal of Respiratory and Critical Care Medicine 2011;183(1 MeetingAbstracts):A5373. [CENTRAL: 1031450] [EMBASE: 70849677] [Google Scholar]
  12. Martinez FJ, Rabe KF, Wouters EFM, Brose M, Goehring U, Fabbri LM, et al. Time course and reversibility of weight decrease with roflumilast, a phosphodiesterase 4 inhibitor. American Journal of Respiratory and Critical Care Medicine 2010;181(1 Meeting Abstracts):A4441. [CENTRAL: 1031630] [EMBASE: 70841891] [Google Scholar]
  13. Nowak D, Ehlken B, Kotchie R, Wecht S, Magnussen H. Roflumilast in combination with long-acting bronchodilators. Deutsche Medizinische Wochenschrift 2013;138(4):119-25. [DOI] [PubMed] [Google Scholar]
  14. Wedzicha JA, Rabe KF, Martinez FJ, Bredenbroker D, Brose M, Goehring UM, et al. Efficacy of roflumilast in the COPD frequent exacerbator phenotype. Chest 2013;143(5):1302-11. [CENTRAL: 870960] [PMID: ] [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐125 {published data only}

  1. Andrew M, Fernando J, HERMES Study Team. Efficacy and safety of the phosphodiesterase 4 inhibitor roflumilast in patients with symptomatic chronic obstructive pulmonary disease in the M2-125 study. Chest 2009;136(4):93S-94. [Google Scholar]
  2. Calverley PM, Rabe KF, Goehring U-M, Kristiansen S, Fabbri LM, Martinez FJ, et al. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009;374(9691):685-94. [DOI] [PubMed] [Google Scholar]
  3. Nowak D, Ehlken B, Kotchie R, Wecht S, Magnussen H. Roflumilast in combination with long-acting bronchodilators. Deutsche Medizinische Wochenschrift 2013;138(4):119-25. [DOI] [PubMed] [Google Scholar]

Roflumilast M2‐127 {published data only}

  1. Chapman KR, McIvor A, Maltais F, EOS Study Team. Additional clinical benefit in patients with chronic obstructive pulmonary disease treated with roflumilast and salmeterol. Chest 2009;136(4):3S-f. [Google Scholar]
  2. Chapman KR, Rabe KF. Efficacy and safety of roflumilast in patients with chronic obstructive pulmonary disease (COPD) concomitantly treated with tiotropium or salmeterol. Primary Care Respiratory Journal 2010;19(2):A12 [44]. [Google Scholar]
  3. Fabbri LM, Calverley PM, Izquierdo-Alonso JL, Bundschuh DS, Brose M, Martinez FJ, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long acting bronchodilators: two randomised clinical trials. Lancet 2009;374(9691):695-703. [DOI] [PubMed] [Google Scholar]
  4. Izquierdo JL, MacNee W, Biermann E, Goehring U-M, McIvor A. The PDE4 inhibitor roflumilast provides additional clinical benefit in COPD patients receiving salmeterol. In: European Respiratory Society Annual Congress; 2009 Sep 12-16; Vienna. 2009.
  5. Martinez F, McIvor A, Brose M, Larsson T, Goehring UM. Benefit of roflumilast therapy added to salmeterol in patients with varying chronic obstructive pulmonary disease severity. Chest 2010;138(4):467A. [Google Scholar]
  6. NCT00313209. Effect of roflumilast on lung function in chronic obstructive pulmonary disease (COPD) patients treated with salmeterol: the EOS study (BY217/M2-127). https://clinicaltrials.gov/show/NCT00313209 .
  7. Sun S, Rennard S, Calverley P, Tourkodimitris S, Rowe P, Creanga D, et al. Effect of roflumilast treatment on dyspnea in patients with chronic obstructive pulmonary disease. Journal of Hospital Medicine 2012;7(Suppl 2):S85-6. [Google Scholar]
  8. Sun S, Rennard S, Calverley P, Tourkodimitris S, Rowe P, Creanga D, et al. Effect of roflumilast treatment on health related quality of life in patients with chronic obstructive pulmonary disease. Journal of Hospital Medicine 2012;7(Suppl 2):S81-2. [Google Scholar]

Roflumilast M2‐128 {published data only}

  1. Chapman KR, Rabe KF. Efficacy and safety of roflumilast in patients with chronic obstructive pulmonary disease (COPD) concomitantly treated with tiotropium or salmeterol. Primary Care Respiratory Journal 2010;19(2):A12 [44]. [Google Scholar]
  2. Fabbri LM, Calverley PM, Izquierdo-Alonso JL, Bundschuh DS, Brose M, Martinez FJ, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long acting bronchodilators: two randomised clinical trials. Lancet 2009;374(9691):695-703. [DOI] [PubMed] [Google Scholar]
  3. Fabbri LM, Martinez FJ, Goehring U-M, Brose M, Lakkis H, Rowe P. Roflumilast treatment with concomitant tiotropium: effect on lung function in severe COPD patients. Journal of General Internal Medicine 2012;27:S303. [CENTRAL: 980891] [EMBASE: 71296919] [Google Scholar]
  4. Paggiaro P, Foden A. Improvements in breathlessness in patients with chronic obstructive pulmonary disease treated with roflumilast and tiotropium. Chest 2009;136(4):3S-g, 4. [Google Scholar]
  5. Rabe K, Paggiaro P, Bernabeu L, Brose M, Geohring U-M, Fabbri L. Roflumilast, a PDE4 inhibitor, improves lung function in patients with COPD treated with tiotropium. In: European Respiratory Society Annual Congress; 2009 Sep 12-16; Vienna. 2009.
  6. Rennard SI, Sun S, Tourkodimitris S, Creanga D, Goehring UM, Bredenbroeker D. Effect of roflumilast treatment added to tiotropium on dyspnea in patients with chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2012;185(Meeting Abstracts):A2261. [Google Scholar]
  7. Wouters EFM, Teichmann P, Brose M, Rabe KF, Fabbri LM. Effects of roflumilast, a phosphodiesterase 4 inhibitor, on body composition in chronic obstructive pulmonary disease. American Journal of Respiratory and Critical Care Medicine 2010;181(Meeting Abstracts):A4473. [Google Scholar]

Roflumilast ROF‐MD‐07(RE2SPOND) {published data only}

  1. Ferguson GT, Rennard SI, Hanania NA, Zhu H, Siddiqui S, Sacks H. Roflumilast treatment in COPD patients taking a fixed-dose combination of long-acting β2 agonist (LABA) and inhaled corticosteroid (ICS): rationale and design of a prospective randomized controlled trial. American Journal of Respiratory and Critical Care Medicine 2012;185(Meeting Abstracts):A2946. [Google Scholar]
  2. Martinez FJ, Calverley PMA, Goehring U-M, Hodge R, Fabbri LM, Rabe KF. Effect of roflumilast on exacerbations in patients with severe COPD and a prior history of hospitalization taking combination therapy. European Respiratory Journal 2015;46:OA482. [Google Scholar]
  3. Martinez FJ, Rabe KF, Sethi S, Pizzichini E, McIvor A, Anzueto A, et al. Effect of roflumilast and inhaled corticosteroid/long-acting beta2-agonist on chronic obstructive pulmonary disease exacerbations (RE(2)SPOND). A randomized clinical trial. American Journal of Respiratory and Critical Care Medicine 2016;194(5):559-67. [PMID: ] [DOI] [PubMed] [Google Scholar]
  4. PER-114-11. A 52-week, double-blind, randomized, placebo-controlled, parallel-group study to evaluate the effect of roflumilast 500 µg on exacerbation rate in subjects with chronic obstructive pulmonary disease (COPD) treated with a fixed-dose combination of long-acting beta agonist and inhaled corticosteroid (LABA/ICS). www.who.int/trialsearch/Trial2.aspx?TrialID=PER-114-11 .
  5. Rennard SI, Martinez FJ, Rabe KF, Sethi S, Pizzichini E, McIvor A, et al. Effect of roflumilast in COPD patients receiving inhaled corticosteroid/long-acting beta2-agonist fixed-dose combination: RE2SPOND rationale and study design. International Journal of Chronic Obstructive Pulmonary Disease 2016;11(1):1921-8. [CENTRAL: 1180201] [EMBASE: 20160624756] [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Rennard SI, Martinez FJ, Sethi S, Zhu H, Haberman R, Zovko E. Effects of roflumilast in COPD patients receiving ICS/LABA fixed-dose combination: rationale and design of a prospective randomized controlled trial. American Journal of Respiratory and Critical Care Medicine 2015;191(Meeting Abstracts):A5790. [CENTRAL: 1101144] [EMBASE: 72053688] [Google Scholar]
  7. Sethi S, Martinez FJ, Rabe KF, Pizzichini E, McIvor A, Anzueto A, et al. Effect of roflumilast on cough and sputum in patients with severe or very severe chronic obstructive pulmonary disease (COPD) receiving inhaled combination therapy: evaluation of the exacerbation of chronic pulmonary disease tool-patient reported outcomes (exact-pro) subdomain scores. American Journal of Respiratory and Critical Care Medicine 2017;195:A1335. [Google Scholar]
  8. White WB, Kowey PR, Zhu H, Siddiqui S, Rowe P. Evaluation of major adverse cardiac events (MACE) in a one-year, placebo-controlled study of roflumilast in patients with chronic obstructive pulmonary disease (COPD): rationale and design. American Journal of Respiratory and Critical Care Medicine 2013;187(Meeting Abstracts):A1484. [CENTRAL: 870804] [Google Scholar]

Urban 2018 (ELASTIC) {published data only}

  1. Urban M, Kreibich N, Funk G-C, Burghuber OC. Effects of roflumilast on subclinical atherosclerosis in COPD - a randomised controlled trial [Effects of the anti-inflammatory drug "ROFLUMILAST" on markers of early atherosclerosis in chronic obstructive pulmonary disease]. clinicaltrialsregister.eu/ctr-search/trial/2011-004152-19/AT (first received 17 February 2012).
  2. Urban M, Kreibich N, Funk G-C, Burghuber OC. Effects of roflumilast on subclinical atherosclerosis in COPD - a randomised controlled trial. European Respiratory Journal 2017;50:PA693. [Google Scholar]

References to studies excluded from this review

Borker 2003 {published data only}

  1. Borker RD, Morris A, Lim J, Zhu J, Reisner C. Effect of cilomilast on quality of life improvement/deterioration and non-drug costs in patients with chronic obstructive pulmonary disease. Chest 2003;124(4):170S-b,171. [Google Scholar]

CTRI/2012/09/002961 {published data only}

  1. CTRI/2012/09/002961. A randomized, open labelled, multi centric parallel group three arms clinical study to evaluate the efficacy and safety of roflumilast 500mcg tablet in chronic obstructive pulmonary disorder. who.int/trialsearch/Trial2.aspx?TrialID=CTRI/2012/09/002961 (first received 5 September 2012).

CTRI/2014/01/004370 {published data only}

  1. CTRI/2014/01/004370. A clinical trial study of oral Roflumilast 0.5mg Tablet and combination therapy of Roflumilast 0.5 mg tablet plus Salmeterol 25mcg oral inhaler and combination therapy of Roflumilast 0.5 mg tablet plus Tiotropium 9mcg oral inhaler in adult patients with chronic obstructive pulmonary disease [An open-label, prospective, three arm, parallel group, randomized, multicentric phase-III clinical study to evaluate the efficacy and safety between monotherapy of oral Roflumilast 0.5mg Tablet and combination therapy of Roflumilast 0.5 mg tablet plus Salmeterol 25mcg oral inhaler and combination therapy of Roflumilast 0.5 mg tablet plus Tiotropium 9mcg oral inhaler in adult patients with chronic obstructive pulmonary disease]. who.int/trialsearch/Trial2.aspx?TrialID=CTRI/2014/01/004370 (first received 31 January 2014).

Ferguson 2003 {published data only}

  1. Ferguson G, Fischer TL, Morris A, Zhu J, Barnhart F, Reisner C. Cardiovascular safety of cilomilast in patients with chronic obstructive pulmonary disease. Chest 2003;124(4):171S. [Google Scholar]

Fischer 2003 {published data only}

  1. Fischer T, Borker R, Barnhart F, Morris A, Zhu J. Effect of cilomilast on chronic obstructive pulmonary disease patients with impaired quality of life. Chest 2003;124(4):129S. [Google Scholar]

Grootendorst 2001 {published data only}

  1. Grootendorst DC, Gauw SA, Kelly J, Murdoch RD, Sterk PJ, Rabe KF. First dose bronchodilating effect of phosphodiesterase-4 (PDE-4) inhibition by cilomilast (Ariflo) with or without co-administration of salbutamol and/or ipratropium in COPD patients. European Respiratory Journal 2001;18(Suppl 33):1:35s. [Google Scholar]

Grootendorst 2002 {published data only}

  1. Grootendorst DC, Gauw SA, Verhoosel R, Van der Veen H, Van der Linden A, Moesker H, et al. Effect of a PDE4 inhibitor (Bay 19-8004) on FEV1 and airway inflammation in patients with COPD. American Journal of Respiratory and Critical Care Medicine 2002;165(8 Suppl):A226. [Google Scholar]

Grootendorst 2003 {published data only}

  1. Grootendorst DC, Gauw SA, Baan R, Kelly J, Murdoch RD, Sterk PJ, et al. Does a single dose of the phosphodiesterase 4 inhibitor, cilomilast (15mg), induce bronchodilation in patients with chronic obstructive pulmonary disease? Pulmonary Pharmacology and Therapeutics 2003;16(2):115-20. [DOI] [PubMed] [Google Scholar]

Grootendorst 2007 {published data only}

  1. Grootendorst DC, Gauw SA, Verhoosel RM, Sterk PJ, Hospers JJ, Bredenbröker D, et al. Reduction in sputum neutrophil and eosinophil numbers by the PDE4 inhibitor roflumilast in patients with COPD. Thorax 2007;62(12):1081-7. [DOI] [PMC free article] [PubMed] [Google Scholar]

GSK256066 {published data only}

  1. Lazaar AL, Mistry S, Barrett C, Lulic-Burns Z. A four-week randomized study of the safety and tolerability of the inhaled PDE4 inhibitor GSK256066 in COPD. American Journal of Respiratory and Critical Care Medicine 2010;181(Meeting Abstracts):A4444. [Google Scholar]

Kelsen 2002 {published data only}

  1. Kelsen SG, Rennard SI, Chodosh S, Schryver B, Vleisides C, Zhu J. COPD exacerbation in a 6-month trial of cilomilast (Ariflo), a potent, selective phosphodiesterase 4 inhibitor. American Journal of Respiratory and Critical Care Medicine 2002;165(Suppl 8):A271. [Google Scholar]

Knobil 2003 {published data only}

  1. Knobil K, Morris A, Zhu J, Fischer T, Reisner C. Cilomilast is efficacious in chronic obstructive pulmonary disease. In: American Thoracic Society 99th International Conference; 2003 May 16-21; Seattle. 2003:A035; Poster D92.
  2. Reisner C, Morris A, Zhu J, Fischer T, Knobil K. Cilomilast is efficacious in chronic obstructive pulmonary disease. European Respiratory Journal 2003;22(Suppl 45):P530. [Google Scholar]

Lim 2004 {published data only}

  1. Lim S, Zhu J, Lake P. Cilomilast decreases exacerbations and maintains lung function in patients with poorly reversible COPD. European Respiratory Journal 2004;24(Suppl 48):88s. [Google Scholar]

NCT00246935 {published data only}

  1. NCT00246935. Long-term study of safety and efficacy of roflumilast in Japanese patients older than 40 years with chronic obstructive pulmonary disease (APTA-2217-08) [A long-term study of APTA-2217 in patients with chronic obstructive pulmonary disease]. clinicaltrials.gov/show/nct00246935 (first received 1 November 2005).

NCT01849341 {published data only}

  1. NCT01849341. Roflumilast safety administered once a day on alternate days for two weeks compared to the usual dosage once daily [Clinical trial phase III blind, parallel group to analyze differences in the safety of roflumilast administered once a day on alternate days for two weeks compared to the usual dosage once daily]. clinicaltrials.gov/show/NCT01849341 (first received 8 May 2013).

NCT01973998 {published data only}

  1. NCT01973998. Effects of roflumilast in hospitalized chronic obstructive pulmonary disease (COPD) on mortality and re-hospitalization. clinicaltrials.gov/show/nct01973998 (first received 1 November 2013).

NCT02018432 {published data only}

  1. NCT02018432. Strategy to improve adherence of roflumilast [Adherence to therapy in COPD patients under dose escalation of roflumilast]. clinicaltrials.gov/show/nct02018432 (first received 23 December 2013).

Nieman 1999 {unpublished data only}

  1. Nieman RB, Taneja DT, Amit O, Benincosa LJ, Compton CH, Bethala VK, et al. The effects of low dose SB207499, a second generation, oral PDE4 inhibitor, in patients with COPD. In: European Respiratory Society Congress; 1999 Oct 9-13; Madrid. 1999.

Pascoe 2007 {unpublished data only}

  1. Pascoe SJ, Bonner J, Hauffe S, Bohnemeier H. Gradual dose escalation of QAK423, a novel PDE4 inhibitor, significantly improves the tolerability. In: American Thoracic Society International Conference; 2007 May 18-23; San Francisco. 2007.

Rabe 2017 {published data only}

  1. Rabe KF, Calverley PMA, Martinez FJ, Fabbri LM. Effect of roflumilast in patients with severe COPD and a history of hospitalisation. European Respiratory Journal 2017;50(1):1700158. [DOI] [PubMed] [Google Scholar]

Reisner 2003 {published data only}

  1. Reisner C, Morris A, Barnhart F, Fischer TL, Acusta A, Darken P. Cilomilast reduces exacerbations in patients with chronic obstructive pulmonary disease. Chest 2003;124:4. [Google Scholar]

Rennard 2008 {published data only}

  1. Rennard S, Knobil K, Rabe KF, Morris A, Schachter N, Locantore N, et al. The efficacy and safety of cilomilast in COPD. Drugs 2008;68(Suppl 2):3-57. [DOI] [PubMed] [Google Scholar]

Roflumilast JP708 {unpublished data only}

  1. Brown P. Center for drug evaluation and research application number: 022522Orig1s000. Pharmacology review. www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022522Orig1s000PharmR.pdf (accessed prior to 28 June 2017).

Sadigov 2014 {published data only}

  1. Sadigov A, Akhundov S, Bagirov R. Analysis of chronic obstructive pulmonary disease exacerbations with the triple therapy compared with dual and single bronchodilator therapy: which treatment is better for patients with severe disease? Chest 2014;145(3):425A. [CENTRAL: 991341] [EMBASE: 71429002] [Google Scholar]
  2. Sadigov AS, Bagirov R, Abbasov C. Analysis of chronic obstructive pulmonary disease exacerbations with the triple therapy compared with dual treatment: is it better treatment tool for patients with severe disease? American Journal of Respiratory and Critical Care Medicine 2014;189:A3770. [CENTRAL: 1035664] [EMBASE: 72043281] [Google Scholar]

Sadigov 2015 {published data only}

  1. Sadigov A, Huseynova S. Efficacy and safety of dual anti-inflammatory combination of fluticasone and roflumilast for the treatment of COPD: is dual better than single? American Journal of Respiratory and Critical Care Medicine 2015;191(Meeting Abstracts):A3968. [CENTRAL: 1101148] [EMBASE: 72051845] [Google Scholar]

SB207499/040 {unpublished data only}

  1. 207499/040. A multicentre, open-label extension study to evaluate the safety, tolerability and efficacy of oral SB-207499 (15 mg twice daily) in patients with chronic obstructive pulmonary disease. www.gsk-clinicalstudyregister.com/files/pdf/24044.pdf (first received 28 September 2008).

SB207499/041 {unpublished data only}

  1. 207499/041. A multicenter open-label extension study to evaluate the safety, tolerability and efficacy of oral cilomilast (15 mg twice daily) in patients with chronic obstructive pulmonary disease. www.gsk-clinicalstudyregister.com/files/pdf/24045.pdf (first received 28 September 2008).

Song 2005 {published data only}

  1. Song Y, Wang C, Liao X, Wang Y, Li Q, Zhao Z, et al. Improvement in lung residual volume in patients with COPD roles of anti-inflammation activity of cilomilast. Respiratory 2005;10(Suppl 3):A135. [Google Scholar]

Spencer 2002 {published data only}

  1. Spencer MD, Zhu J, Izard D. The direct costs of exacerbations in COPD and the effect of cilomilast treatment. European Respiratory Journal 2002;20(Suppl 38):245s. [Google Scholar]

Vestbo 2007 {published data only}

  1. Vestbo J, Tan L, Atkinson G. A 6 week study of the efficacy and safety of UK 500,001 dry powder for inhalation (DPI) in adults with chronic obstructive pulmonary disease. European Respiratory Journal 2007;30(Suppl 51):612s [P3598]. [Google Scholar]

Vestbo 2009 {published data only}

  1. Vestbo J, Tan L, Atkinson G, Ward J. A controlled trial of 6-weeks' treatment with a novel inhaled phosphodiesterase type-4 inhibitor in COPD. European Respiratory Journal 2009;33(5):1039-44. [DOI] [PubMed] [Google Scholar]

Wang 2005 {published data only}

  1. Wang C, Song Y, Liao X. Efficacy and anti-inflammation activity of a selective phospodiesterase-4 inhibitor cilomilast in treatment of COPD. Chest 2005;128(4):262S-a. [Google Scholar]

Watz 2013 {published data only}

  1. Watz H, Mistry SJ, Lazaar AL, IPC101939 investigators. Safety and tolerability of the inhaled phosphodiesterase 4 inhibitor GSK256066 in moderate COPD. Pulmonary Pharmacology and Therapeutics 2013;26(5):588-95. [CENTRAL: 872117] [EMBASE: 2013527752] [PMID: ] [DOI] [PubMed] [Google Scholar]

Watz 2016 {published data only}

  1. EUCTR2013-001788-21-SK. Study to evaluate how to optimise the use of roflumilast in subjects who have a lung disease called chronic obstructive pulmonary disease (COPD) [A multicenter, randomized, double-blind phase 3 study to evaluate tolerability and pharmacokinetics of 500 µg roflumilast once daily with an up-titration regimen in COPD, including an open-label down-titration period evaluating tolerability and pharmacokinetics of 250 µg roflumilast once daily in subjects not tolerating 500 µg roflumilast once-daily]. clinicaltrialsregister.eu/ctr-search/trial/2013-001788-21/results (first received 6 February 2014).
  2. Facius A, Bagul N, Gardiner P, Watz H. Pharmacokinetics and pharmacodynamics of a 4-week up-titration regimen of roflumilast in the optimize study OS. Pneumologie 2018;72(Suppl 1):S92. [Google Scholar]
  3. Facius, Bagul N, Gardiner P, Watz H. Pharmacokinetics of a 4-week up-titration regimen of roflumilast in the optimize study. American Journal of Respiratory and Critical Care Medicine. 2017;195:A1337. [Google Scholar]
  4. NCT02165826. Evaluation of tolerability and pharmacokinetics of roflumilast, 250μg and 500μg, as add-on to standard COPD treatment to treat severe COPD (OPTIMIZE). clinicaltrials.gov/ct2/show/NCT02165826 (first received 18 June 2014).
  5. Watz H, Bagul N, Nip K, Sun R, Goehring U-M, Calverley P, et al. Tolerability of different dosing regimens of roflumilast in severe COPD (OPTIMIZE). European Respiratory Journal 2016;48:PA308. [Google Scholar]
  6. Watz H, Bagul N, Rabe KF, Rennard S, Alagappan VKT, Roman J, et al. Use of a 4-week up-titration regimen of roflumilast in patients with severe COPD. International Journal of Chronic Obstructive Pulmonary Disease 2018;13:813-22. [DOI] [PMC free article] [PubMed] [Google Scholar]

References to studies awaiting assessment

Barnes 2014 {published data only}

  1. Barnes NC, Saetta M, Rabe KF. Implementing lessons learned from previous bronchial biopsy trials in a new randomized controlled COPD biopsy trial with roflumilast. BMC Pulmonary Medicine 2014;14(1):9. [CENTRAL: 973300] [EMBASE: 2014126619] [PMID: ] [DOI] [PMC free article] [PubMed] [Google Scholar]

EUCTR2004‐004442‐40‐GB {published data only}

  1. EUCTR2004-004442-40-GB. 500µg roflumilast once daily in combination with 50µg salmeterol twice daily versus 50µg salmeterol twice daily alone over 52 weeks in patients with COPD. who.int/trialsearch/Trial2.aspx?TrialID=EUCTR2004-004442-40-GB (first received 23 February 2005).

Mahmud 2013 {published data only}

  1. Mahmud AM, Hossain A, Hassan R, Khan AS, Bennoor KS, Shaheen M, et al. Placebo controlled study of roflumilast in Bangladeshi COPD patients. Respirology 2013;18(Suppl 4):125 [PS160]. [CENTRAL: 980913] [EMBASE: 71371785] [Google Scholar]

NCT00671073 {published data only}

  1. NCT00671073. Study to assess efficacy and safety of oglemilast in patients with chronic obstructive pulmonary disease (COPD). clinicaltrials.gov/show/NCT00671073 (first received 2 May 2008).

NCT01595750 {published data only}

  1. NCT01595750. Randomized, double-blind, placebo-controlled study to evaluate the effect of roflumilast on endothelial function in patients with chronic obstructive pulmonary disease (REVASC) [Randomized, double-blind, placebo-controlled study to evaluate the effect of roflumilast on endothelial function in patients with chronic obstructive pulmonary disease]. clinicaltrials.gov/show/NCT01595750 (first received 10 May 2012).

NCT01701934 {published data only}

  1. NCT01701934. Impact of roflumilast on visceral adiposity and metabolic profile in chronic obstructive pulmonary disease (RAMBO). clinicaltrials.gov/show/NCT01701934 (first received 5 October 2012).

References to ongoing studies

NCT02451540 2015 {published data only}

  1. 2015-000053-21. Placebo controlled study to assess the effect of Roflumilast in hyperinflated COPD patients in addition to LABA/LAMA therapy using functional respiratory imaging. clinicaltrialsregister.eu/ctr-search/trial/2015-000053-21/BE (first received 14 April 2015).
  2. NCT02451540. Evaluation of the effect of roflumilast in hyperinflated COPD patients using functional respiratory imaging [Placebo controlled study to assess the effect of roflumilast in hyperinflated COPD patients in addition to LABA/LAMA therapy using functional respiratory imaging]. clinicaltrials.gov/show/NCT02451540 (first received 7 May 2015).

NCT02671942 2016 {published data only}

  1. NCT02671942. A multicenter randomized double-blind clinical study evaluated the safety, pharmacokinetic and pharmacodynamic characteristics of roflumilast in COPD patients. clinicaltrials.gov/show/NCT02671942 (first received 25 January 2016).

Additional references

Agusti 2005

  1. Agusti A. COPD, a multicomponent disease: implications for management. Respiratory Medicine 2005;99(6):670-82. [DOI] [PubMed] [Google Scholar]

Barnes 2000

  1. Barnes P. Medical progress: chronic obstructive pulmonary disease. New England Journal of Medicine 2000;343:269-80. [DOI] [PubMed] [Google Scholar]

Barnes 2003

  1. Barnes P. Theophylline: new perspectives for an old drug. American Journal of Respiratory and Critical Care Medicine 2003;167(6):813-8. [DOI] [PubMed] [Google Scholar]

Barnes 2005

  1. Barnes P. Theophylline in chronic obstructive pulmonary disease: new horizons. Proceedings of the American Thoracic Society 2005;2(4):334-9. [DOI] [PubMed] [Google Scholar]

Boswell‐Smith 2006

  1. Boswell-Smith V, Spina D, Page C. Phosphodiesterase inhibitors. British Journal of Pharmacology 2006;147:s252-7. [DOI] [PMC free article] [PubMed] [Google Scholar]

Calverley 2007

  1. Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. New England Journal of Medicine 2007;356(8):775-89. [DOI] [PubMed] [Google Scholar]

Calverley 2009

  1. Calverley PM, Rabe KF, Goehring UM, Kristiansen S, Fabbri LM, Martinez FJ. Roflumilast in symptomatic chronic obstructive pulmonary disease: two randomised clinical trials. Lancet 2009;374:685-94. [DOI] [PubMed] [Google Scholar]

Celli 2004

  1. Celli B, MacNee W. Standards for the diagnosis and treatment of patients with COPD: a summary of the ATS/ERS position paper. European Respiratory Journal 2004;23(6):932-46. [DOI] [PubMed] [Google Scholar]

Cochrane Airways 2019

  1. Cochrane Airways Trials Register. airways.cochrane.org/trials-register (accessed 7 May 2019).

Donohue 2005

  1. Donohue J. Minimal clinically important differences in COPD lung function. COPD 2005;2:111-24. [DOI] [PubMed] [Google Scholar]

Essayan 2001

  1. Essayan D. Cyclic nucleotide phosphodiesterases. Journal of Allergy and Clinical Immunology 2001;108(5):671-80. [DOI] [PubMed] [Google Scholar]

Fabbri 2009

  1. Fabbri LM, Calverley PM, Izquierdo-Alonso JL, Bundschuh DS, Brose M, Martinez FJ, et al. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with long acting bronchodilators: two randomised clinical trials. Lancet 2009;374(9691):695-703. [DOI] [PubMed] [Google Scholar]

GOLD 2020

  1. From the global strategy for the diagnosis, management and prevention of COPD, global initiative for chronic obstructive lung disease (GOLD) 2020. goldcopd.org (accessed 11 February 2020).

GRADEpro GDT 2015 [Computer program]

  1. McMaster University (developed by Evidence Prime) GRADEpro GDT. Version accessed 31 May 2017. Hamilton (ON): McMaster University (developed by Evidence Prime), 2015.Available at gradepro.org.

Guyatt 2008

  1. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, Schünemann HKJ. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. British Medical Journal 2008;336:924. [DOI] [PMC free article] [PubMed] [Google Scholar]

Higgins 2003

  1. Higgins JT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327:557-60. [DOI] [PMC free article] [PubMed] [Google Scholar]

Higgins 2019

  1. Higgins JT, Savović J, Page MJ, Elbers RG, Sterne AC. Chapter 8. Assessing risk of bias in a randomized trial. In: Higgins JT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (editors). Cochrane Handbook for Systematic Reviews of Interventions version 6.0 (updated July 2019). Cochrane, 2019. Available from www.training.cochrane.org/handbook.

Jones 2005

  1. Jones P. St. George's Respiratory Questionnaire: MCID. COPD 2005;2:75-9. [DOI] [PubMed] [Google Scholar]

Le Rouzic 2018

  1. Le Rouzic O, Roche N, Cortot AB, Tillie-Leblond I, Masure F, Perez T et al. Defining the "frequent exacerbator" phenotype in COPD. Chest Journal 2018;153(5):1106-15. [DOI] [PubMed] [Google Scholar]

Luo 2016

  1. Luo J, Wang K, Liu D, Liang BM, Liu CT. Can roflumilast, a phosphodiesterase-4 inhibitor, improve clinical outcomes in patients with moderate-to-severe chronic obstructive pulmonary disease? A meta-analysis. Respiratory Research 2016;17:17. [DOI] [PMC free article] [PubMed] [Google Scholar]

Mathers 2005

  1. Mathers C, Loncar D. Updated projections of global mortality and burden of disease, 2002-2030: data sources, methods and results. Evidence and Information for Policy Working Paper. who.int/healthinfo/statistics/bod_projections2030_paper.pdf (accessed prior to 28 June 2017).

Moher 2009

  1. Moher D, Liberati A, Tetzlaff J, Altman DG, the PRISMA Group (2009). Preferred Reporting Items for Systematic Reviews and Meta-Analyses: The PRISMA Statement. PLoS Medicine 6;7:e1000097. [DOI: 10.1371/journal.pmed1000097] [DOI] [PMC free article] [PubMed] [Google Scholar]

Rennard 2014

  1. Rennard SI, Sun SX, Tourkodimitris S, Rowe P, Goehring UM, Bredenbröker D, et al. Roflumilast and dyspnea in patients with moderate to very severe chronic obstructive pulmonary disease: a pooled analysis of four clinical trials. International Journal of Chronic Obstructive Pulmonary Disease 2014;9:657-73. [DOI] [PMC free article] [PubMed] [Google Scholar]

RevMan 2014 [Computer program]

  1. Nordic Cochrane Centre, The Cochrane Collaboration Review Manager 5 (RevMan 5). Version 5.3. Copenhagen: Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Shen 2018

  1. Shen LF, Lv XD, Chen WY, Yang Q, Fang ZX, Lu WF. Effect of roflumilast on chronic obstructive pulmonary disease: a systematic review and meta-analysis. Irish Journal of Medical Science 2018;187(3):731-8. [DOI] [PubMed] [Google Scholar]

TORCH 2007

  1. Calverley P, Anderson J, Celli B, Ferguson GT, Jenkins C, Jones P, et al. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. New England Journal of Medicine 2007;356(8):775-89. [DOI] [PubMed] [Google Scholar]

Torphy 1998

  1. Torphy T. Phosphodiesterase isozymes: molecular targets for novel antiasthma agents. American Journal of Respiratory and Critical Care Medicine 1998;157(2):351-70. [DOI] [PubMed] [Google Scholar]

Torphy 1999

  1. Torphy T, Barnette M, Underwood D, Griswold DE, Christensen SB, Murdoch RD, et al. Ariflo (SB 207499), a second generation phosphodiesterase 4 inhibitor for the treatment of asthma and COPD: from concept to clinic. Pulmonary Pharmacology and Therapeutics 1999;12(2):131-5. [DOI] [PubMed] [Google Scholar]

Van Geffen 2015

  1. Van Geffen WH, Slebos DJ, Kerstjens HA. Hyperinflation in COPD exacerbations. Lancet Respiratory Medicine 2015;12:e43-44. [DOI] [PubMed] [Google Scholar]

Vignola 2004

  1. Vignola A. PDE4 inhibitors in COPD - a more selective approach to treatment. Respiratory Medicine 2004;98(6):495-503. [DOI] [PubMed] [Google Scholar]

Wedzicha 2007

  1. Wedzicha JA, Seemungal TA. COPD exacerbations: defining their cause and prevention. Lancet 2007;370:786-96. [DOI] [PMC free article] [PubMed] [Google Scholar]

Wedzicha 2016

  1. Wedzicha JA, Calverley PMA, Rabe KF. Roflumilast: a review of its use in the treatment of COPD. International Journal of Chronic Obstructive Pulmonary Disease 2016;11:81-90. [DOI] [PMC free article] [PubMed] [Google Scholar]

White 2003

  1. White AJ, Gompertz S, Stockley RA. Chronic obstructive pulmonary disease. 6: the aetiology of exacerbations of chronic obstructive pulmonary disease. Thorax 2003;58:73-80. [DOI] [PMC free article] [PubMed] [Google Scholar]

White 2013

  1. White W, Cooke G, Kowey P, Calverley P, Bredenbröker D, Goehring U, et al. Cardiovascular safety in patients receiving roflumilast for the treatment of chronic obstructive pulmonary disease. Chest 2013;144(3):758-65. [DOI] [PubMed] [Google Scholar]

Yuan 2016

  1. Yuan L, Dai X, Yang M, Cai Q, Shao N. Potential treatment benefits and safety of roflumilast in COPD: a systematic review and meta-analysis. International Journal of Chronic Obstructive Pulmonary Disease 2016;11:1477-83. [DOI] [PMC free article] [PubMed] [Google Scholar]

References to other published versions of this review

Chong 2013

  1. Chong J, Leung B, Poole P. Phosphodiesterase 4 inhibitors for chronic obstructive pulmonary disease. Cochrane Database of Systematic Reviews 2013, Issue 11. [DOI: 10.1002/14651858.CD002309.pub4] [DOI] [PubMed] [Google Scholar]

Articles from The Cochrane Database of Systematic Reviews are provided here courtesy of Wiley

RESOURCES