1,925
Views
6
CrossRef citations to date
0
Altmetric
Lung Health Workshop

Stable COPD Treatment: Where are We?

Pages 123-129 | Received 05 Feb 2018, Accepted 20 Feb 2018, Published online: 02 Apr 2018

ABSTRACT

The number of pharmacological treatments available for COPD has increased markedly in the last years, mostly corresponding to new agents, combinations and devices within know pharmacological classes. Hierarchizing these options is not straightforward since expected effects are limited by the intrinsically fixed character of the underlying lung damage. In addition, all options have not been directly compared face-to-face. Therefore, guidelines derive from some level of subjective interpretation of the available evidence. Determining which magnitude of change can be taken as clinically relevant is complex although crucial to define long-term strategies. Similarly, estimating not only the possible benefits but also the risks of treatments at the individual level is of major importance to guide choices. In the future biomarkers may be of help in that respect. They will hopefully emerge from progresses in systems biology and medicine. Before then, prescriptions should be restricted to the appropriate treatment indications, as established by high level studies and formalized by guidelines.

Introduction

Significant changes in the conceptual framework of guidelines on COPD treatment have occurred during the last decade, following the results of large therapeutic trials (Citation1–4). The purpose of this opinion paper is not to report an extensive analysis of this literature, but to provide the reader with some clues to understand and interpret current trends in COPD management. As a result, a limited number of references has been chosen arbitrarily to illustrate the messages, without any attempt at being exhaustive. Before considering any therapy, every caregiver shall keep in mind that the first and most universal measures to implement when initiating COPD management are smoking cessation interventions, physical activity counselling and vaccination against influenza and Streptococcus pneumoniae. Risk reduction also includes measures against occupational or domestic exposures, when indicated (Citation3). With this in mind, the present article will focus on pharmacological treatments, which should not prevent clinicians from considering the important roles of rehabilitation (indicated in ALL patients in whom some exercise limitation remains despite optimal pharmacological therapy) and, in very specific subgroups, instrumental support (oxygen therapy and noninvasive ventilation) and interventional lung volume reduction (Citation3). Of note, the Cochrane collaboration decided to stop performing systematic reviews and meta-analyses on the beneficial effects of rehabilitation versus conventional care in terms of quality of life (Citation5). This illustrates that clinical research on this topic has already given the best possible level of evidence, although the most optimal modalities remain to be explored in more details.

Goals of COPD management and target mechanisms

A major question to address when designing treatment guidelines relates to the definition of goals of care. In the COPD area, these have been clearly stated, e.g., in the GOLD document, and include symptoms relief (i.e., decreased dyspnea, improved exercise tolerance and quality of life) and prevention of future risks (i.e., risks of exacerbation, disease progression and premature death). Reaching these goals could be considered as some form of disease control. However, although this term (“control”) has a precise significance in asthma, it does not correspond to a well-defined notion in COPD yet (Citation6).

Once treatment goals are defined, knowledge of the mechanisms underlying the impact of disease will allow identifying potential treatment targets. For instance, relieving dyspnea requires to reduce the level of hyperinflation through bronchodilation while fighting muscle deconditioning, thereby acting on the two most important components of the vicious circles that link lung alterations to impaired quality of life and handicap () (Citation7). Reducing exacerbations requires to target inflammation and infection (Citation8), but more detailed insights in underlying targets remain necessary. For instance, the role of the microbiome and its modulation represent an area of intensive research (Citation9).

Figure 1. Mechanisms linking chronic airflow limitation to quality of life impairment. Adapted from (Citation7) with permission.

Figure 1. Mechanisms linking chronic airflow limitation to quality of life impairment. Adapted from (Citation7) with permission.

A benefit-risk approach to select the best pharmacological options

Pharmacological approaches comprise short-acting (SA) and long-acting (LA) bronchodilators and agents with anti-inflammatory properties. Bronchodilators belong to two classes, i.e., beta2 agonists (BA) and anticholinergic (AC) agents. Anti-inflammatory drugs include inhaled corticosteroids (ICS), phosphodiesterase-4 inhibitors (PDE4i), macrolides (azithromycin), mucoactive agents with antioxidant effects. Theophylline has both bronchodilator and anti-inflammatory properties, but its therapeutic index is narrow, limiting its use due to side-effects and multiple interactions with food, smoking and other treatments.

The choice between these various agents needs to rely on a benefit-risk approach based on the individual patient‘s characteristics (Citation10): possible benefits will derive from the patient's disease manifestations (mainly dyspnea, exacerbations and their correlates in terms of exercise tolerance, handicap, quality of life) and the possible treatment effects as documented through randomized clinical trials and eventually supported by observational evidence. Possible risks will be deduced from the patient's individual risk factors and comorbidities ().

Figure 2. Components of the benefit-risk balance of current COPD treatments. Adapted from (Citation10) with permission.

Figure 2. Components of the benefit-risk balance of current COPD treatments. Adapted from (Citation10) with permission.

Inhaled treatments have long been considered as completely safe since they aim at delivering high amounts of drugs locally with minimal systemic exposure. However, there is evidence from randomized controlled trials and/or observational studies that inhaled corticosteroids administered at high doses (those authorized for most products in COPD) during many years have the potential to induce lung infections (pneumonia, tuberculosis, nontuberculous mycobacteria, aspergillosis) and systemic side-effects (Citation11–14). Regarding long-acting bronchodilators, a recent large database study has suggested an increase of cardiovascular events following their initiation (Citation15). Even if these analyses do not correspond to the highest possible level of evidence, they are sufficient to induce caution, underlining that the prescription of these agents should be restricted to their recognized indications.

Determining the best strategy, from levels of evidence to clinical judgment

Current guideline documents rely mostly on symptoms (dyspnea) and exacerbations to define treatment indications. Some variations do exist between guidelines although all are based on the same evidence. These differences illustrate the weight of interpretation in the development of recommendations. A major question when considering treatment effects is to determine their clinical relevance. The magnitude of the effects (i.e., of the differences between the considered treatment and placebo on the one hand, active comparators on the other) must be put in the perspective of the impact of the disease and its intrinsically limited reversibility. Other aspects to consider include the availability, cost and practicality of treatments as well as patients' preferences and abilities, which are crucial in the field of inhaled therapy: in COPD as in asthma, adherence to treatment and inhalation technique are poor and tightly associated with “disease control” (Citation16,Citation17) which encompasses symptoms burden and exacerbation risk (Citation6).

With all the advantages and limitations of each treatment option in mind, the challenge is to determine whether therapeutic strategies should systematically follow a step-by-step approach, or whether some categories of patients deserve maximal treatment from the beginning when the disease is diagnosed at a relatively advanced stage: in other words, should a newly diagnosed patient with a mMRC dyspnea grade of 3, three acute respiratory episodes in the past year and a FEV1 at 42% predicted (i.e., GOLD D category and GOLD 3 level of airflow limitation) be treated with a long-acting bronchodilator, a dual combination (i.e., a LABA+LAMA or a LABA+ICS) or triple therapy (ICS+LABA+LAMA) right from the beginning? No study has assessed the respective benefits of these approaches in initially naïve patients, making it impossible to provide strictly evidence-based answers to this question. Therefore, the choice will result from the estimation of the added value of one strategy over the other, based on differences between available options: e.g., is it worth initiating treatment with a combination of agents that provide a 10% absolute increase in the proportion of responders in terms of dyspnea, from 40% to 50%, when compared to a single agent? Or should the prescriber keep in mind that 80% of those who would respond to the combination would also respond to the single agent? Similarly, is a 15% decrease in exacerbation frequency with a dual approach compared to mono-component sufficient to justify dual therapy as first-line option? This issue is complexified by the variability of exacerbation frequency at an individual level (Citation18) despite the statistically significant value of past exacerbation rate for prediction of future exacerbations at a group level (Citation19). This is where subjectivity plays a major role, explaining why hours of discussions are often required within guidelines committees to produce an apparently simple (although often challenged subsequently) recommendation based on apparently strong randomized trials. To help clinical decision-making, many studies have been performed to define the minimal clinically important differences for variables used to assess treatment efficacy (Citation20). Various methods have been used, based on anchoring strategies and distribution analyses. Interpreting their results is complex since they can be influenced by many factors including disease severity, comparator (active agent or placebo), poor relations between clinical and lung function variables, concomitant therapy and comorbidities, context of care… The relatively limited responsiveness to treatments overall (as opposed to asthma, for instance) is another point to consider.

Network analyses where direct comparisons are lacking

Another challenge is the appraisal of possible differences between agents within a given family, when face-to-face comparisons are mostly performed with lung function rather than clinical outcomes as a primary endpoint. Extrapolation from these studies is made difficult by the poor relation between lung function and clinical improvements. As a result, it is also difficult to determine whether results obtained with a given agent or combination of agents can be extrapolated to other drugs of the same family(ies). Thus, network meta-analyses have been performed to allow indirect comparisons between agents that have not been directly compared in terms of clinical endpoints, through the combination of studies with a common comparator (Citation21). However, although the methodology of these analyses is now better standardized, it seems difficult to ensure that patients from different studies are sufficiently similar to compare treatment effects with full confidence: subtle differences in respiratory characteristics, comorbidities, previous and concomitant treatments, disease history may induce masked residual confounding.

Guidelines paradigms and a selection of the underlying evidence

Current guidelines are more explicitly based on patients' categorization than their ancestors. Decision trees are extensively used, some guidelines aiming at presenting one single algorithm for all clinical situations () (Citation22–25) while others propose specific algorithms for specific patients' populations () (Citation3). Altogether, differences between these documents are rather minimal, although their presentations vary (Citation26). The main results on which these recommendations are based are the following: for exacerbation prevention, LAMAs do better than LABAs; (Citation27) ICS+LABA combinations are superior to their mono-components in that respect (Citation28–30), but less effective than LABA+LAMA combinations, at least in patients with no history of asthma and less than 600 eosinophils/µL (Citation31). Bronchodilator combinations are more efficacious than mono-components in terms of lung function and clinical endpoints (Citation32), which could relate to not only additive but also synergistic effects resulting from receptor cross-talk (Citation33). However, this may not translate in improved exercise capacity, due to peripheral limitations related to skeletal muscle dysfunction (Citation34). More recent studies have explored the potential of triple combinations of LABA+LAMA+ICS, especially regarding exacerbation prevention (Citation35–37). Superiority of triple therapy has been demonstrated versus LAMA alone and LABA+ICS. Results addressing the comparison with LABA+LAMA should be published soon. In the WISDOM trial, patients have been treated with triple therapy for a few weeks before being randomly allocated between ICS withdrawal (leaving them on LABA+LAMA) and ICS continuation (leaving them on triple therapy) (Citation38). No increase in exacerbations occurred in the withdrawal arm, although some lung function was lost. However, since this was a step-down study, it does not allow to drive conclusions as to the differences between LABA+LAMA and triple therapy when prescribed in a step-up approach.

Figure 3. Propositions of the French-Language Respiratory Society for COPD pharmacological treatment. Adapted from (Citation22) with permission.

Figure 3. Propositions of the French-Language Respiratory Society for COPD pharmacological treatment. Adapted from (Citation22) with permission.

Figure 4. GOLD recommendations for COPD pharmacological treatment. Adapted from (Citation3) with permission.

Figure 4. GOLD recommendations for COPD pharmacological treatment. Adapted from (Citation3) with permission.

Importantly, interpreting the results of randomized trials requires evaluating how they can be extrapolated to the general COPD population. Unfortunately, there is still a paucity of high-level real-life research in the field of COPD pharmacological treatment. Only very few pragmatic trials have been performed (Citation39) and rigorous observational research remains limited. Although these study designs can certainly not replace randomized controlled trials they may be of help to generate hypotheses especially regarding the best target populations and the most appropriate strategies regarding device choices (Citation40). In that sense, they can be of use to inform guidelines (Citation41), especially considering that patients recruited in randomized clinical trials represent only a minority of the COPD patients population, although this proportion has improved over time (Citation42,43).

Towards (more) precision medicine

Large cohort studies have been used to identify clinical COPD phenotypes (Citation44) associated with relevant outcomes and, possibly, specific treatment effects (Citation19,Citation45,Citation46). However, in most therapeutic studies subgroup analyses aiming at identifying responders have provided disappointing results, preventing from identifying specific target subgroups. One exception is the PDE4i roflumilast, which is specifically indicated in patients with severe airflow obstruction, chronic bronchitis and exacerbations despite long-acting bronchodilators (Citation47). Long-term treatment with azithromycin may be more effective at preventing exacerbations in older former smokers with mild airflow obstruction, but this requires prospective testing (Citation48).

The lack of sufficient predictive ability of clinical characteristics to identify treatment responders has triggered increasing interest in the possible role of biomarkers to guide treatment choices. In particular, increased blood eosinophils (although remaining within the normal range) appear associated with a greater chance of response to inhaled corticosteroids in terms of exacerbation reduction (Citation49). Indeed, at least in some patients (identified through their blood eosinophil count), the role of eosinophils in COPD pathophysiology is strongly suggested by the efficacy of anti-IL-5 agents for exacerbation prevention (Citation50). However, more studies remain necessary to confirm prospectively that eosinophil-based strategies can be recommended and determine the best thresholds and the most adequate target populations, especially since blood eosinophil counts are somehow variable within individuals and merely reflect airway eosinophilia (Citation51). Progresses in systems/network biology and medicine, made possible by advances in bioinformatics (Citation52) and the development of biological and clinical databases, should open new avenues for translational research and therapeutic developments (Citation53).

To conclude, although at its early stages, precision COPD care is already there. It will hopefully progress in the coming years, allowing more targeted therapeutic choices with more favorable benefit-risk profiles at the individual patient level. However, the lung damages observed in COPD prevent lung function abnormalities from being markedly reversible with current treatment options. Regenerative medicine may contribute to reverse these damages (Citation54). Awaiting such advances, guidelines implementation needs to be improved to avoid current deviations that appear frequent (Citation55,Citation56) and may be associated with suboptimal patients' outcomes. This requires to better understand the factors involved in the way guidelines affect real-life prescriptions (Citation57). Another important component of guidelines on pharmacological therapy should be a check-list of what needs to be done before changing prescribed medicines () (Citation22).

Table 1. Check list of questions to address before modifying pharmacological treatment for COPD. Adapted from (Citation22) with permission.

Disclosure

Dr. Roche reports grants and personal fees from Boehringer Ingelheim, Pfizer and Novartis, personal fees from Teva, GSK, AstraZeneca, Chiesi, Mundipharma, Cipla, Sanofi, Sandoz, 3M, Zambon, outside the submitted work.

References

  • Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd SS. Global strategy for the diagnosis, management, and prevention of chronic obstructive pulmonary disease. NHLBI/WHO Global Initiative for Chronic Obstructive Lung Disease (GOLD) Workshop summary. Am J Respir Crit Care Med. 2001;163(5):1256–76. doi:10.1164/ajrccm.163.5.2101039. PMID:11316667.
  • Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A, Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, et al. Global strategy for the diagnosis, management and prevention of chronic obstructive pulmonary disease, GOLD executive summary. Am J Respir Crit Care Med. August 2012. doi:10.1164/rccm.201204-0596PP.
  • Vogelmeier CF, Criner GJ, Martinez FJ, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Chen R, Decramer M, Fabbri LM, et al. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease 2017 report: GOLD executive summary. Eur Respir J. 2017;49(3). doi:10.1183/13993003.00214-2017.
  • Rabe KF, Hurd S, Anzueto A, Barnes PJ, Buist SA, Calverley P, Fukuchi Y, Jenkins C, Rodriguez-Roisin R, Van Weel C, et al. Global strategy for the diagnosis, management, and prevention of COPD – 2006 Update. Am J Respir Crit Care Med. 2007;176:532–55. doi:10.1164/rccm.200703-456SO. PMID:17507545.
  • Lacasse Y, Cates CJ, McCarthy B, Welsh EJ. This cochrane review is closed: deciding what constitutes enough research and where next for pulmonary rehabilitation in COPD. Cochrane Database Syst Rev. 2015;(11):ED000107. doi:10.1002/14651858.ED000107. PMID:26593129.
  • Nibber A, Chisholm A, Soler-Cataluña JJ, Alcazar B, Price D, Miravitlles M. Validating the concept of COPD control: A real-world cohort study from the United Kingdom. COPD. 2017;14(5):504–12. doi:10.1080/15412555.2017.1350154. PMID:28813176.
  • Aguilaniu B, Roche N. The difficulties of measuring and improving physical activity in COPD. NPJ Prim Care Respir Med. 2014;24:14014. doi:10.1038/npjpcrm.2014.14. PMID:24841568.
  • Wedzicha JA, Brill SE, Allinson JP, Donaldson GC. Mechanisms and impact of the frequent exacerbator phenotype in chronic obstructive pulmonary disease. BMC Med. 2013;11:181. doi:10.1186/1741-7015-11-181. PMID:23945277.
  • Faner R, Sibila O, Agustí A, Bernasconi E, Chalmers JD, Huffnagle GB, Manichanh C, Molyneaux PL, Paredes R, Pérez Brocal V, et al. The microbiome in respiratory medicine: current challenges and future perspectives. Eur Respir J. 2017;49(4):1602086. doi:10.1183/13993003.02086-2016. PMID:28404649.
  • Woodruff PG, Agusti A, Roche N, Singh D, Martinez FJ. Current concepts in targeting chronic obstructive pulmonary disease pharmacotherapy: making progress towards personalised management. Lancet. 2015;385(9979):1789–98. doi:10.1016/S0140-6736(15)60693-6. PMID:25943943.
  • Price D, Yawn B, Brusselle G, Rossi A. Risk-to-benefit ratio of inhaled corticosteroids in patients with COPD. Prim Care Respir J. 2013;22(1):92–100. doi:10.4104/pcrj.2012.00092. PMID:23135217.
  • Bulpa P, Bihin B, Dimopoulos G, Taccone FS, Van den Abeele A-M, Misset B, Meersseman W, Spapen H, Cardoso T, Charles P-E, et al. Which algorithm diagnoses invasive pulmonary aspergillosis best in ICU patients with COPD? Eur Respir J. 2017;50(3):1700532. doi:10.1183/13993003.00532-2017. PMID:28954783.
  • Andréjak C, Nielsen R, Thomsen VØ, Duhaut P, Sørensen HT, Thomsen RW. Chronic respiratory disease, inhaled corticosteroids and risk of non-tuberculous mycobacteriosis. Thorax. 2013;68(3):256–62. doi:10.1136/thoraxjnl-2012-201772. PMID:22781123.
  • Dong Y-H, Chang C-H, Lin Wu F-L, Shen L-J, Calverley PMA, Löfdahl C-G, Lai M-S, Mahler DA. Use of inhaled corticosteroids in patients with COPD and the risk of TB and influenza: A systematic review and meta-analysis of randomized controlled trials. a systematic review and meta-analysis of randomized controlled trials. Chest. 2014;145(6):1286–97. doi:10.1378/chest.13-2137. PMID:24504044.
  • Wang M-T, Liou J-T, Lin CW, Tsai C-L, Wang Y-H, Hsu Y-J, Lai J-H. Association of cardiovascular risk with inhaled long-acting bronchodilators in patients with chronic obstructive pulmonary disease: a nested case-control study. JAMA Intern Med. 2018;178(2):229–38. doi:10.1001/jamainternmed.2017.7720. PMID:29297057.
  • Melani AS, Bonavia M, Cilenti V, Cinti C, Lodi M, Martucci P, Serra M, Scichilone N, Sestini P, Aliani M, et al. Inhaler mishandling remains common in real life and is associated with reduced disease control. Respir Med. 2011;105(6):930–8. doi:10.1016/j.rmed.2011.01.005. PMID:21367593.
  • Molimard M, Raherison C, Lignot S, Balestra A, Lamarque S, Chartier A, Droz-Perroteau C, Lassalle R, Moore N, Girodet P-O. Chronic obstructive pulmonary disease exacerbation and inhaler device handling: real-life assessment of 2935 patients. Eur Respir J. 2017;49(2):1601794. doi:10.1183/13993003.01794-2016. PMID:28182569.
  • Han MK, Quibrera PM, Carretta EE, Barr RG, Bleecker ER, Bowler RP, Cooper CB, Comellas A, Couper DJ, Curtis JL, et al. Frequency of exacerbations in patients with chronic obstructive pulmonary disease: an analysis of the SPIROMICS cohort. Lancet Respir Med. 2017;5(8):619–26. doi:10.1016/S2213-2600(17)30207-2. PMID:28668356.
  • Hurst JR, Vestbo J, Anzueto A, Locantore N, Müllerova H, Tal-Singer R, Miller B, Lomas DA, Agusti A, Macnee W, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363(12):1128–38. doi:10.1056/NEJMoa0909883. PMID:20843247.
  • Jones PW, Beeh KM, Chapman KR, Decramer M, Mahler DA, Wedzicha JA. Minimal clinically important differences in pharmacological trials. Am J Respir Crit Care Med. 2014;189(3):250–5. doi:10.1164/rccm.201310-1863PP. PMID:24383418.
  • Donohue JF, Betts KA, Du EX, Altman P, Goyal P, Keininger DL, Gruenberger J-B, Signorovitch JE. Comparative efficacy of long-acting β2-agonists as monotherapy for chronic obstructive pulmonary disease: a network meta-analysis. Int J Chron Obstruct Pulmon Dis. 2017;12:367–81. doi:10.2147/COPD.S119908. PMID:28176892.
  • Zysman M, Chabot F, Housset B, Morelot Panzini C, Devillier P, Roche N, Société de Pneumologie de Langue Française. Pharmacological treatment optimisation for stable COPD: an endless story? Proposals from the Société de Pneumologie de Langue Française. Eur Respir J. 2017;50(4). doi:10.1183/13993003.01250-2017. PMID:29025880.
  • Aisanov Z, Avdeev S, Arkhipov V, Belevskiy A, Chuchalin A, Leshchenko I, Ovcharenko S, Shmelev E, Miravitlles M. Russian guidelines for the management of COPD: algorithm of pharmacologic treatment. Int J Chron Obstruct Pulmon Dis. 2018;13:183–7. doi:10.2147/COPD.S153770. PMID:29386887.
  • Kankaanranta H, Harju T, Kilpeläinen M, Mazur W, Lehto JT, Katajisto M, Peisa T, Meinander T, Lehtimäki L. Diagnosis and pharmacotherapy of stable chronic obstructive pulmonary disease: the finnish guidelines. Basic Clin Pharmacol Toxicol. 2015;116(4):291–307. doi:10.1111/bcpt.12366. PMID:25515181.
  • Miravitlles M, Anzueto A. A new two-step algorithm for the treatment of COPD. Eur Respir J. 2017;49(2):1602200. doi:10.1183/13993003.02200-2016. PMID:28179443.
  • Miravitlles M, Vogelmeier C, Roche N, Halpin D, Cardoso J, Chuchalin AG, Kankaanranta H, Sandström T, Śliwiński P, Zatloukal J, et al. A review of national guidelines for management of COPD in Europe. Eur Respir J. 2016:47(2):625–37. doi:10.1183/13993003.01170-2015. PMID:26797035.
  • Vogelmeier C, Hederer B, Glaab T, Schmidt H, Rutten-van Mölken MPMH, Beeh KM, Rabe KF, Fabbri LM. Tiotropium versus salmeterol for the prevention of exacerbations of COPD. N Engl J Med. 2011;364(12):1093–103. doi:10.1056/NEJMoa1008378. PMID:21428765.
  • Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Yates JC, Vestbo J. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med. 2007;356(8):775–89. doi:10.1056/NEJMoa063070. PMID:17314337.
  • Szafranski W, Cukier A, Ramirez A, Menga G, Sansores R, Nahabedian S, Peterson S, Olsson H. Efficacy and safety of budesonide/formoterol in the management of chronic obstructive pulmonary disease. Eur Respir J. 2003;21(1):74–81. doi:10.1183/09031936.03.00031402. PMID:12570112.
  • Singh D, Kampschulte J, Wedzicha JA, Jones PW, Cohuet G, Corradi M, Higenbottam T, Petruzzelli S, Vestbo J. A trial of beclomethasone/formoterol in COPD using EXACT-PRO to measure exacerbations. Eur Respir J. 2013;41(1):12–7. doi:10.1183/09031936.00207611. PMID:22653766.
  • Wedzicha JA, Banerji D, Chapman KR, Vestbo J, Roche N, Ayers RT, Thach C, Fogel R, Patalano F, Vogelmeier CF, et al. Indacaterol-glycopyrronium versus salmeterol-fluticasone for COPD. N Engl J Med. 2016;374(23):2222–34. doi:10.1056/NEJMoa1516385. PMID:27181606.
  • Bateman ED, Chapman KR, Singh D, D'Urzo AD, Molins E, Leselbaum A, Gil EG. Aclidinium bromide and formoterol fumarate as a fixed-dose combination in COPD: pooled analysis of symptoms and exacerbations from two six-month, multicentre, randomised studies (ACLIFORM and AUGMENT). Respir Res. 2015;16:92. doi:10.1186/s12931-015-0250-2. PMID:26233481.
  • Cazzola M, Calzetta L, Segreti A, Facciolo F, Rogliani P, Matera MG. Translational study searching for synergy between glycopyrronium and indacaterol. COPD. 2015;12(2):175–81. doi:10.3109/15412555.2014.922172. PMID:25222881.
  • Beeh K-M, Korn S, Beier J, Jadayel D, Henley M, D'Andrea P, Banerji D. Effect of QVA149 on lung volumes and exercise tolerance in COPD patients: the BRIGHT study. Respir Med. 2014;108(4):584–92. doi:10.1016/j.rmed.2014.01.006. PMID:24534204.
  • Singh D, Papi A, Corradi M, Pavlišová I, Montagna I, Francisco C, Cohuet G, Vezzoli S, Scuri M, Vestbo J. Single inhaler triple therapy versus inhaled corticosteroid plus long-acting β2-agonist therapy for chronic obstructive pulmonary disease (TRILOGY): a double-blind, parallel group, randomised controlled trial. Lancet. 2016;388(10048):963–73. doi:10.1016/S0140-6736(16)31354-X. PMID:27598678.
  • Vestbo J, Papi A, Corradi M, Blazhko V, Montagna I, Francisco C, Cohuet G, Vezzoli S, Scuri M, Singh D. Single inhaler extrafine triple therapy versus long-acting muscarinic antagonist therapy for chronic obstructive pulmonary disease (TRINITY): a double-blind, parallel group, randomised controlled trial. Lancet 2017;389(10082):1919–29. doi:10.1016/S0140-6736(17)30188-5. PMID:28385353.
  • Lipson DA, Barnacle H, Birk R, Brealey N, Locantore N, Lomas DA, Ludwig-Sengpiel A, Mohindra R, Tabberer M, Zhu C-Q, et al. FULFIL trial: once-daily triple therapy for patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2017;196(4):438–46. doi:10.1164/rccm.201703-0449OC. PMID:28375647.
  • Magnussen H, Disse B, Rodriguez-Roisin R, Kirsten A, Watz H, Tetzlaff K, Towse L, Finnigan H, Dahl R, Decramer M, et al. Withdrawal of inhaled glucocorticoids and exacerbations of COPD. N Engl J Med. 2014;371(14):1285–94. doi:10.1056/NEJMoa1407154. PMID:25196117.
  • Vestbo J, Leather D, Diar Bakerly N, New J, Gibson JM, McCorkindale S, Collier S, Crawford J, Frith L, Harvey C, et al. Effectiveness of fluticasone furoate-vilanterol for COPD in clinical practice. N Engl J Med. 2016;375(13):1253–60. doi:10.1056/NEJMoa1608033. PMID:27593504.
  • Bosnic-Anticevich S, Chrystyn H, Costello RW, Dolovich MB, Fletcher MJ, Lavorini F, Rodríguez-Roisin R, Ryan D, Wan Yau Ming S, Price DB. The use of multiple respiratory inhalers requiring different inhalation techniques has an adverse effect on COPD outcomes. Int J Chron Obstruct Pulmon Dis. 2017;12:59–71. doi:10.2147/COPD.S117196. PMID:28053517.
  • Roche N, Reddel HK, Agusti A, Bateman ED, Krishnan JA, Martin RJ, Papi A, Postma D, Thomas M, Brusselle G, et al. Integrating real-life studies in the global therapeutic research framework. Lancet Respir Med. 2013;1(10):e29–30. doi:10.1016/S2213-2600(13)70199-1. PMID:24461762.
  • Halpin DMG, Kerkhof M, Soriano JB, Mikkelsen H, Price DB. Eligibility of real-life patients with COPD for inclusion in trials of inhaled long-acting bronchodilator therapy. Respir Res. 2016;17(1):120. doi:10.1186/s12931-016-0433-5. PMID:27663386.
  • Herland K, Akselsen J-P, Skjønsberg OH, Bjermer L. How representative are clinical study patients with asthma or COPD for a larger “real life” population of patients with obstructive lung disease? Respir Med. 2005;99(1):11–19. doi:10.1016/j.rmed.2004.03.026. PMID:15672843.
  • Han MK, Agusti A, Calverley PM, Celli BR, Criner G, Curtis JL, Fabbri LM, Goldin JG, Jones PW, Macnee W, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182(5):598–604. doi:10.1164/rccm.200912-1843CC. PMID:20522794.
  • Burgel P-R, Paillasseur J-L, Roche N. Identification of clinical phenotypes using cluster analyses in COPD patients with multiple comorbidities. BioMed Res Int. 2014;2014:420134. doi:10.1155/2014/420134. PMID:24683548.
  • Garcia-Aymerich J, Gómez FP, Benet M, Farrero E, Basagaña X, Gayete À, Paré C, Freixa X, Ferrer J, Ferrer A, et al. Identification and prospective validation of clinically relevant chronic obstructive pulmonary disease (COPD) subtypes. Thorax. 2011;66(5):430–7. doi:10.1136/thx.2010.154484. PMID:21177668.
  • 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:10.1016/S0140-6736(14)62410-7. PMID:25684586.
  • Han MK, Tayob N, Murray S, Dransfield MT, Washko G, Scanlon PD, Criner GJ, Casaburi R, Connett J, Lazarus SC, et al. Predictors of chronic obstructive pulmonary disease exacerbation reduction in response to daily azithromycin therapy. Am J Respir Crit Care Med. 2014;189(12):1503–8. doi:10.1164/rccm.201402-0207OC. PMID:24779680.
  • Bafadhel M, Pavord ID, Russell REK. Eosinophils in COPD: just another biomarker? Lancet Respir Med. 2017;5:747–759. doi:10.1016/S2213-2600(17)30217-5.
  • Pavord ID, Chanez P, Criner GJ, Kerstjens HAM, Korn S, Lugogo N, Martinot J-B, Sagara H, Albers FC, Bradford ES, et al. Mepolizumab for eosinophilic chronic obstructive pulmonary disease. N Engl J Med. 2017;377(17):1613–29. doi:10.1056/NEJMoa1708208. PMID:28893134.
  • Roche N, Zysman M, Burgel P-R. Blood eosinophil counts as a guide for COPD treatment strategies. Lancet Respir Med. January 2018. doi:10.1016/S2213-2600(18)30005-5.
  • Agustí A, Celli B, Faner R. What does endotyping mean for treatment in chronic obstructive pulmonary disease? Lancet 2017;390:980–7. doi:10.1016/S0140-6736(17)32136-0. PMID:28872030.
  • Agusti A, Gea J, Faner R. Biomarkers, the control panel and personalized COPD medicine. Respirol Carlton Vic. 2016;21(1):24–33. doi:10.1111/resp.12585.
  • Oh DK, Kim Y-S, Oh Y-M. Lung regeneration therapy for chronic obstructive pulmonary disease. Tuberc Respir Dis. 2017;80(1):1–10. doi:10.4046/trd.2017.80.1.1.
  • Vestbo J, Vogelmeier C, Small M, Higgins V. Understanding the GOLD 2011 Strategy as applied to a real-world COPD population. Respir Med. 2014;108(5):729–36. doi:10.1016/j.rmed.2014.03.002. PMID:24675239.
  • Burgel P-R, Deslée G, Jebrak G, Brinchault G, Caillaud D, Chanez P, Court-Fortune I, Escamilla R, Nesme-Meyer P, Paillasseur J-L, et al. Real-life use of inhaled corticosteroids in COPD patients versus the GOLD proposals: a paradigm shift in GOLD 2011? Eur Respir J. 2014;43(4):1201–3. doi:10.1183/09031936.00162313. PMID:24176996.
  • Boulet L, Becker A, Bowie D, McIvor A, Hernandez P, Rouleau M, Bourbeau J, Graham I, Legare F, Ward T, et al. Implementing practice guidelines: a workshop on guidelines dissemination and implementation with a focus on asthma and COPD. Can Respir J. 2006;13(Suppl A):5–47. doi:10.1155/2006/810978. PMID:16552449.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.