4,148
Views
18
CrossRef citations to date
0
Altmetric
Review

Inhaled Corticosteroids for Chronic Obstructive Pulmonary Disease—The Shifting Treatment Paradigm

, &

Abstract

Chronic obstructive pulmonary disease (COPD) guidelines suggest using inhaled corticosteroids (ICS) in patients with severe airflow limitation or those at high risk of exacerbations. This recommendation is based on evidence demonstrating that ICS, especially when prescribed in fixed-dose combinations (FDC) with long-acting β2 agonists (LABA), improve quality of life (QoL), decrease exacerbations and hospitalisations, and have been associated with a trend towards a reduction in all-cause mortality. Audit shows that routine prescribing practice frequently uses inhaler therapies outside current guidelines recommendations; severe to very severe disease constitutes about 20% of all COPD patients, but up to 75% of COPD patients are prescribed an ICS, with significant numbers given ICS/LABA as first-line maintenance therapy. The role of ICS in the treatment paradigm for COPD is changing, driven by the growing evidence of increased risk of pneumonia, and the introduction of a new class of FDC; LABA and long-acting muscarinic antagonists (LAMA), which simplify dual bronchodilation and present a plausible alternative therapy. As the evidence base for dual therapy bronchodilation expands, it is likely that maximal bronchodilation will move up the treatment algorithm and ICS reserved for those with more severe disease who are not controlled on dual therapy. This change has already manifested in local COPD algorithms, such as those at Tayside, and represents a significant change in recommended prescribing practice. This review reassesses the role of ICS in the shifting treatment paradigm, in the context of alternative treatment options that provide maximal bronchodilation.

Abbreviations
COPD=

chronic obstructive pulmonary disease

FDC=

fixed-dose combinations

FEV1=

forced expiratory volume in one second

GOLD=

Global Initiative for Chronic Obstructive Lung Disease

ICS=

inhaled corticosteroids

INSPIRE=

Investigating New Standards for Prophylaxis in Reduction of Exacerbations

LABA=

long-acting beta agonist

LAMA=

long-acting muscarinic antagonist

MRC=

Medical Research Council

NICE=

National Institute for Health and Clinical Excellence

NNH=

number needed to harm

NNT=

number needed to treat

QoL=

quality of life

SABA=

short-acting β2-agonist

SAMA=

short-acting muscarinic antagonist

SAL=

salmeterol

SFC=

salmeterol/fluticasone propionate

SGRQ=

St Georges Respiratory Questionnaire

TDI=

transition dyspnoea index

TORCH=

Towards a Revolution in COPD Health

Introduction

Important changes in our perspective and understanding of chronic obstructive pulmonary disease (COPD) have led to marked improvements in the treatment approach (Citation1). Disease management has moved beyond spirometric parameters to include patient centred outcomes such as dyspnoea and exacerbation frequency, which are recognised as important elements in the assessment of treatment effectiveness, and there is acknowledgment of the importance of COPD phenotyping in optimising therapeutic options. In 2013, the Global Initiative for Chronic Obstructive Lung Disease (GOLD) issued an updated document detailing the classification and treatment of COPD (Citation1).

GOLD recommends classifying patients into one of four risk groups (A-D) using a combination of spirometry, symptoms and risk of future exacerbations. Treatment algorithms were suggested for each group, however bronchodilator therapy has remained the cornerstone of pharmacological ­management in all groups. Indeed, long-acting muscarinic antagonists (LAMA) are a first-line treatment option for any but those with mildest disease. GOLD suggests considering the use of inhaled corticosteroids (ICS) in patients with severe airflow limitation or for those who are at high risk of exacerbations. This is based on evidence demonstrating that ICS, especially when prescribed in fixed-dose combinations (FDC) with long-acting β2 agonists (LABA), improve quality of life (QoL), decrease exacerbations, hospitalisations (Citation2), and have been associated with a trend towards a reduction in all-cause mortality (Citation3). Dual bronchodilation, through co-prescribing of LABA and LAMA, could be an alternative to ICS/LABA in certain COPD phenotypes. Also this approach could be used particularly in patients who remain symptomatic after LABA monotherapy, but who cannot tolerate or do not wish to take ICS. This concept, however, is not given the prominence it merits in the new GOLD, (Citation1) or indeed other, guidance (Citation1, Citation4).

The pivotal study which showed benefits of ICS/LABA, recruited participants with a forced expiratory volume in one second (FEV1) of less than 60% predicted (Citation3). There is no evidence supporting the therapeutic benefit of ICS/LABA in COPD patients with less severe airflow limitation, irrespective of exacerbation history (Citation5). Despite this, ICS prescribing patterns demonstrate far wider application in COPD pharmacotherapy than recommended, with ICS/LABA a widely prescribed maintenance drug. Audit shows that routine prescribing practice frequently uses inhaled therapies outside current guideline recommendations; severe to very severe disease constitutes about 20% of all COPD patients, but up to 75% of COPD patients are prescribed an ICS, with significant numbers given ICS/LABA as first-line drug (Citation6). This prescribing pattern may reflect the familiarity of prescribing ICS/LABA including historical prescribing where drug therapy is maintained because of disease stability, but it may also be a manifestation of physician concerns over the accuracy of diagnosis of COPD versus asthma resulting in continued ICS/LABA use.

The breadth of prescribing of ICS in COPD and lack of efficacy data in individuals with a FEV1 > 60% highlights the need for the risk-benefit profile of ICS to be considered closely. ICS/LABA combinations are associated with a number of side effects beyond the topical effects of oral candidiasis and dysphonia, the most serious of which is an increased incidence of pneumonia (Citation7). Randomised controlled trials and observational studies have persistently described the risk of pneumonia associated with ICS use, especially with fluticasone (Citation3, Citation8). It has been suggested that there is an intra-class difference in ICS with respect to the risk of pneumonia; however, this is not a consistent observation (Citation9). In addition, recent clinical trial data has suggested that the pneumonia extends to a new ICS currently in development (Citation10, 11).

The evidence base for the efficacy and safety of dual bronchodilation has been limited until now (Citation12). However the evidence for new LABA/LAMA FDC suggests a greater efficacy than LABA or LAMA monotherapy (Citation13). This greater efficacy has been shown over a diverse range of clinically relevant outcomes, and identifies LABA/LAMA FDC as a potential new treatment option when trying to maximise symptom control and reduce exacerbations. This review reassesses the role of ICS in the shifting treatment paradigm, in the context of alternative treatment options that provide maximal bronchodilation.

Review

Inhaled corticosteroids in chronic obstructive pulmonary disease

Thirty years ago, ICS were established as effective treatments to reduce morbidity and mortality in asthma patients (Citation14). This success prompted research into its possible therapeutic role in COPD, although initial evidence of efficacy was limited. The randomised controlled trials of ICS in COPD which followed in the 1990's had mixed results. There was no improvement in the rate of decline in lung function or exacerbation frequency when compared with placebo in studies of 6 months to 3 years duration (Citation5, Citation15–19). It was not until ISOLDE (Citation20); the first large, long-term, randomised, double-blind, placebo-controlled trial of ICS, that reduction in exacerbation rates and health care utilisation of ICS as therapy was described. From this point on, clinical trials focused on assessing ICS/LABA combinations as well as ICS and LABA as monotherapy.

TRISTAN was one of the first, large (N = 1465), double-blind, randomised studies, which compared twice-daily salmeterol/fluticasone propionate 50/500 μg (SFC) to salmeterol 50 μg (SAL), fluticasone propionate 500 μg (FP) or placebo (Citation21). Although the primary endpoint was pre-bronchodilator FEV1, the number of exacerbations was an important secondary endpoint assessed at every clinic visit. All therapies improved lung function, symptoms, health status and reduced use of rescue medication and frequency of exacerbations compared to placebo; however combination therapy with SFC was more effective than salmeterol alone. The treatment effect was most pronounced in severe disease; those with a baseline FEV1 < 50% showed a 30% reduction in exacerbation frequency with SFC compared to placebo, whereas there was only a 10% reduction in those patients whose FEV1 > 50%.

The seminal TORCH trial (Towards a Revolution in COPD Health) followed several years later; this randomised 6112 patients to SFC (100/1000 μg/day), SAL (100 μg/day), FP (1000 μg/day) or placebo (Citation3). All-cause mortality was the primary endpoint, with lung function, exacerbations and QoL reported over a period of 3 years. The SFC treatment group demonstrated, a 25% reduction in the annual rate of exacerbations (0.85 (95% CI, 0.80 to 0.90)) compared to placebo (1.13 (95% CI, 1.07 to 1.20)), with a number needed to treat (NNT) of four to prevent one exacerbation. At the same time, Kardos and colleagues compared SFC with SAL monotherapy in 994 patients with severe COPD for around 4 years (Citation22). Combination therapy with SFC reduced the total number of exacerbations (334 vs. 464, p < 0.0001), the annual rate of moderate/severe exacerbations (0.92 vs. 1.4, corresponding to a 35% decrease, p < 0.0001) and the mean time to first exacerbation (128 vs. 93 days, p < 0.0001) (Citation22).

A significant limitation of the studies described above is that they recruited patients based at least in part on an arbitrary FEV1 cut off rather than the underlying phenotype ie emphysema or chronic bronchitis; it is well established that ICS will have beneficial effects in those with predominantly moderate degrees of airway obstruction and airway hyperresponsiveness or modest reversibility of FEV1 (Citation23). A lack of phenotyping is also present when it comes to exacerbations, many of the studies looking into the usefulness of ICS tend to simply group all exacerbations together, however this may limit a study's power to demonstrate ICS effectiveness. COPD exacerbations can be very different: infective (viral and/or bacterial), eosinophilic predominant or pauci-inflammatory (Citation24). There is evidence that different treatments can be effectively adopted to reduce different COPD exacerbations, for instance, ICS added to long-acting bronchodilators can be utilised effectively to avoid eosinophilic exacerbations (Citation25).

Summary evidence of the effect of ICS on COPD exacerbations is provided from recent Cochrane meta-analyses (Citation2, Citation26). Analysis of 55 primary studies totalling 16,154 participants comparing any dose of any type of ICS with a placebo control in patients with COPD, showed long-term use of ICS significantly reduced the mean rate of exacerbations (generic inverse variance analysis: MD −0.26 exacerbations/patient/year, pooled means analysis: MD −0.19 exacerbations/patient/year)(Citation20). In a pooled analysis of 5601 participants from 6 studies with predominantly poorly reversible, severe COPD, the use of ICS/LABA FDC significantly reduced exacerbation rates (rate-ratio 0.87, 95% CI 0.80 to 0.94) compared with ICS alone (Citation26).

In terms of lung function and QoL, post-hoc analyses of TORCH reported an adjusted rate of decline in FEV1 of 55 mL/year for placebo, 42 mL/year with FP or SAL, and for the combination of drugs a 39 mL per year decline, suggesting that treatment with ICS, LABA or the combination of two, can slow FEV1 decline (Citation27). The Cochrane analysis reported a pooled difference in rate of decline of post-bronchodilator FEV1 of 6.88 mL/year (95% CI 1.80 to 11.96, 5 studies, 4823 participants) in favour of ICS (Citation2). TORCH reported an improvement with SFC of 3.0 units of the St Georges Respiratory Questionnaire (SGRQ) compared to baseline after 3 years of treatment (p < 0.001 vs. placebo), with an improvement of 1.8 units in the fluticasone group (p < 0.001 vs. placebo); the placebo group showed a deterioration of 0.2 units in the SGRQ. The effect was largest in more severe COPD—an improvement in SGRQ of 5.9 units with combination therapy in stage 4, and 3.3 units with GOLD stage 3; in stage 2, the SGRQ improvement was less (2.3 units), but still significant (Citation3). Despite these positive findings, the prescribing of ICS for COPD has remained a controversial subject (Citation28, 29)

Safety profile of ICS treatment

The side effects of ICS treatment range from frequent unpleasant local side effects, such as oral candidiasis and dysphonia (Citation2), to less common ones, such as adrenal suppression (Citation30), cataracts (Citation31, 32), and pneumonia (Citation3, Citation8, Citation32–35). The side effect profile potentially extends to include osteoporotic fractures (Citation2, Citation36) and increased ­diabetes risk (Citation37, 38).

The studies of osteoporosis and fracture risk include combinations of randomised controlled trials and observational studies, each of which employed different definitions and therefore should be interpreted with caution. Several meta-analyses have reported conflicting results; Loke and colleagues found a modest but statistically significant increased likelihood of fractures in those treated with ICS (Citation36); however, a Cochrane review reported that long-term studies which measured bone effects generally showed no major effect on fractures and bone mineral density over 3 years of follow-up (Citation2).

Systemic corticosteroids are known to increase diabetes risk, but the impact of ICS is less well characterised. A large population based study of 388,584 patients with respiratory disease found treatment with ICS was ­associated with a 34% (RR 1.34 (95% CI 1.29–1.39)) increased risk of new onset diabetes (defined by initiation of an oral hypoglycaemic agent), with the highest dose associated with the greatest risk (Citation37). More recently, a retrospective study of administrative claims data from the Australian Government Department of Veterans’ Affairs of more than 18,000 patients with diabetes, found there to be an increased risk of diabetes-related hospitalisations with the use of high-dose ICS (Citation38). However, it is the increased risk of pneumonia which represents the greatest concern in regards to the use of ICS in COPD management.

ICS and pneumonia risk

Patients with COPD are at an increased risk of pneumonia due to the nature of the disease itself and risks may be further increased with ICS use. TORCH showed an increased rate of pneumonia among all patients receiving treatment containing ICS, with a two-fold higher rate of pneumonia compared with patients in the placebo arm (Citation3), with a number-needed-to-harm (NNH) of 17 (Citation33). A subsequent analysis of the TORCH data showed that the risk factors for ICS-associated pneumonia were increasing age, worsening spirometry, a history of COPD exacerbations in the year prior to the study, worse Medical Research Council (MRC) dyspnoea scores and a low body mass index; thus not dissimilar to the usual risk factors for pneumonia (Citation39). The subsequent INSPIRE study (Investigating New Standards for Prophylaxis in Reduction of Exacerbations) comparing salmeterol plus fluticasone propionate 50/500 μg twice daily with tiotropium 18 μg once daily, reported double the rate of pneumonia with SFC (8%) than with tiotropium (4%) (Citation34). The evidence of a dose-response for pneumonia risk comes from a nested case-control study of 175,906 patients, which found that the highest doses of ICS were associated with the highest rate-ratio for hospitalisation (rate-ratio of 2.25) (Citation35). Summary evidence of the risk is provided by a Cochrane meta-analysis of 14 studies combining 11,794 patients, which reported moderate-level evidence of an increased pneumonia risk (by 50%) with ICS/LABA combinations versus LABA alone; however, mortality was identical between the treatment groups (Citation23).

Randomised controlled studies of new agents currently in clinical development have also reported pneumonia. ILLUMINATE, a double-blind, double-dummy trial in 523 patients with GOLD stages II and III, without exacerbations in the previous year, compared once-daily LABA/LAMA FDC (QVA149 110/50 μg) or twice-daily SFC 50/500 μg for 26 weeks (Citation40). Radiologically confirmed pneumonia was only reported in the SFC treatment group (four patients; 1·5%). A new ICS/LABA FDC, recently approved by the United States Food and Drug Administration (FDA) for the treatment of COPD—fluticasone furoate/vilanterol (FF/VI)—was assessed for exacerbation prevention in COPD patients with a history of exacerbations in two replicate, phase III randomised double-blind trials of 1622 patients (Citation10). Three doses of fluticasone furoate were used (50 μg, 100 μg, and 200 μg) in combination with vilanterol 25 μg. Pneumonia and fractures were reported more frequently with FF/VI than with vilanterol alone. Eight deaths from pneumonia in the FF/VI groups compared with none in the vilanterol only group prompted discussion highlighting that the safety of ICS in COPD is a serious problem (Citation11). The study was not powered to look at differences in pneumonia-specific mortality and overall mortality was not significantly different between the two groups. Interpreting the excess pneumonic episodes is complicated by the fact that ICS/LABA use in COPD decreases the rate of exacerbations and hospitalisations in clinical studies, and that no pneumonia deaths were identified secondary to SFC in TORCH (Citation39). Reassuringly, a study looking at the impact of ICS use on outcome in COPD patients admitted with pneumonia did not show an association with worse mortality or the development of complications (Citation41).

It has been suggested that ICS type may influence the risk, with some studies such as PATHOS suggesting that fluticasone carries a higher risk than others such as budesonide (Citation8, Citation42). In this observational, retrospective cohort study, budesonide/formoterol or SFC were compared for pneumonia event rates, admission to hospital related to pneumonia, and mortality associated with pneumonia. SFC patients were significantly more likely to experience pneumonia and had a higher mortality related to pneumonia than patients treated with budesonide/formoterol. The authors offered plausible explanations for the observed difference, suggesting that the known immunosuppressant potency of fluticasone (Citation10-fold higher than that of budesonide with regard to ex vivo inhibition of human alveolar macrophage innate immune response to bacterial triggers) could explain the findings. It must be noted that this was an observational study and that other studies of budesonide and mometasone in COPD have shown an increased rate of pneumonia associated with use of these agents (Citation9, Citation43). The definitive answer as to whether this phenomenon is a class effect or one specific to fluticasone remains unclear.

ICS prescribing patterns

ICS's important but targeted role in COPD treatment is not reflected in prescribing patterns. In a large Italian study investigating whether pulmonologists follow GOLD recommendations when prescribing treatment for COPD, ICS alone and ICS/LABA combinations were over-prescribed in GOLD stages I and II, in patients without exacerbations in 40% and 64%, respectively. Overall, 62.1% of prescribing was not in keeping with GOLD recommendations (Citation44). Similar findings were described in a French study, which found that more than half of all patients in GOLD class I and II were prescribed ICS (Citation45).

Tayside Allergy and Respiratory Disease Information System (TARDIS) is a structured management programme for patients with COPD (Citation46). Patients are reviewed no less than once annually, and data on spirometry, exacerbations, and prescriptions, along with other variables is collected and recorded. A TARDIS study into the safety of β-blockers in COPD offered valuable insights into real life ICS prescribing patterns in Scotland. In this dataset of 5977 patients, the mean post-bronchodilator FEV1 in patients receiving ICS as monotherapy was 65.3% and 63.1% in those patients receiving ICS/LABA FDC (Citation47).

NICE (National Institute for Health and Clinical Excellence) COPD guideline recommend considering ICS/LABA FDC after short-acting β2 agonists (SABA) or short-acting muscarinic antagonists (SAMA) in patients with FEV1 < 50% predicted who have exacerbations or persistent breathlessness, and as a therapeutic option for those who have persistent exacerbations or breathlessness despite maintenance therapy with a LABA, irrespective of FEV1 (Citation4). According to GOLD, ICS/LABA is appropriate for patients with GOLD FEV1 < 50% or in those at high risk of future exacerbations (Citation1).

Clearly, ICS/LABA remains a valuable treatment option for selected patients with severe COPD or frequent exacerbations, however, phenotyping individuals to direct ICS therapy to those most likely to benefit is an important aim. As described above, COPD is a heterogeneous disease with clinically relevant phenotypes and these emerging phenotypes must be considered when planning therapeutic intervention. For example there is probably small benefit of prescribing high dose ICS to a COPD patient with predominant emphysema especially in the presence of relatively little airflow limitation while potentially exposing the individual to the risk of well recognized adverse side effects while at the same time ICS may potentially be used beneficially albeit without strict licensing criteria in non-smokers or ex-smokers with a relatively preserved FEV1 but with symptoms secondary to recurrent eosinophilic COPD exacerbations.

Despite this, the current possibility of guideline concordant ICS prescription irrespective of FEV1 moves ahead of the evidence, as the proven benefit of ICS is restricted to those with more severe airflow restriction. This is sustained in the opinion of the Scottish Medicines Consortium, which has restricted the license of ICS/LABA to individuals with a FEV1 <50% (Citation48). With emerging data on LABA and LAMA co-prescribing, both as free agents and FDC, the recommendation to consider dual bronchodilation only if ICS prescribing is not appropriate, will need to be altered in future guidelines. Important questions for guideline development groups to address will include establishing the role of dual bronchodilation in treatment algorithms in relation to ICS/LABA. This will involve only identifying which patients are most likely to benefit from dual bronchodilation and also taking into account factors such as risk and impact of side effects, symptoms and patient preference.

Subtle changes to the COPD guideline in Tayside were recently introduced to reflect current treatment options. ICS/LABA FDC are still recommended for persistent breathlessness or repeated exacerbations despite bronchodilator therapy (see step 4 in Table ) with the proviso that ICS/LABA should only be prescribed if there are persistent symptoms and/or exacerbations despite maximal bronchodilator therapy (defined as co-prescribing of LABA and LAMA). In essence, LABA and LAMA co-prescribing has been ‘pulled up’ the treatment algorithm ahead of ICS/LABA, encouraging prescribing of ICS in patients with the most severe ­disease.

Table 1.  COPD treatment algorithm employed by NHS Tayside. The algorithm recommends that a combined inhaled steroid and long-acting bronchodilator should be prescribed only if the patient has persistent breathlessness or repeated exacerbations despite optimal bronchodilator therapy (defined in step 3 as LABA and LAMA co-prescribing, coloured blue) and should be discontinued if no benefit after 4 weeks

Combining LABA and LAMA in COPD

The rationale for co-prescribing of LABA and LAMA is clear; combining long-acting bronchodilators with distinct, complementary and possibly synergistic mechanisms of action, has the potential to maximise bronchodilator response. Muscarinic receptor antagonists inhibit bronchoconstriction by inhibiting the binding of acetylcholine to muscarinic receptors on airway smooth muscle. β2-agonists directly activate β2-adrenoreceptors, causing airway smooth muscle relaxation. Therefore, by simultaneously addressing both mechanisms of bronchodilation, one can potentially modulate against the inter-patient and intra-patient variability in response to individual agents (Citation49). This should lead to a bronchodilator response which would not otherwise be seen if one or other agent were prescribed alone. Clinical studies have shown this potentially synergistic effect of co-prescribing, in which the combined treatment effect of LABA and LAMA is greater, or longer-lasting than expected from single agent bronchodilation. This may relate to M3-receptor blockade by LAMA amplifying and prolonging LABA-induced β2-adrenoreceptors activation (Citation50), and provides mechanistic support for the potential of LABA and LAMA inhaled combinations.

At least 12 randomised clinical trials have set out to investigate clinical outcomes after LABA and LAMA co-prescribing (formoterol, arformoterol, or indacaterol co-prescribed with tiotropium) (Citation51). The largest of these, INTRUST-1 and INTRUST-2 trials, randomised 1134 and 1142 patients with moderate to severe COPD respectively, already receiving open-label tiotropium 18 μg, to indacaterol 150 μg once-daily or placebo for 12 weeks (Citation52). Concurrent use of indacaterol plus tiotropium provided superior bronchodilation compared with tiotropium alone, increasing the FEV1 AUC by 130 and 120 mL, trough by 80 and 70 mL (p < 0.001) and trough IC by 130 and 100 mL (p < 0.01). Importantly, the adverse event (AE) profile was similar between treatments except cough, which was more common in the LABA plus LAMA patients. Meta-analysis of eight randomised trials of tiotropium plus formoterol, compared with tiotropium alone, including 1868 patients with stable COPD, found that treatment with tiotropium plus formoterol improved lung function (weighted mean difference in FEV1 of 105 mL) and symptom scores (weighted mean difference in transitional dyspnoea index (TDI) of 1.5). However, there was no significant reduction in exacerbations seen when compared to treatment with tiotropium alone (Citation53). There was a trend towards a decrease in adverse events, although this was not statistically significant.

LABA and LAMA fixed-dose combinations

The development of a LABA/LAMA FDC is a simple way of ensuring that medication burden is reduced (single inhaler vs. multiple inhalers) thus minimising the risk of non-compliance. The challenge in the development of FDC inhalers is ensuring the balance between drug efficacy and side effect profile. This includes determining the minimally effective doses of LABA's and LAMA's in combination preparations, accepting that they may not be the same as in monotherapy preparations due to combination pharmacodynamics.

A number of LABA/LAMA FDC are currently in clinical development (Table ). One combination, known as QVA149 (Ultibro®, Novartis), a FDC of indacaterol (110 μg) and glycopyrronium (50 μg), is the first once-daily dual agent bronchodilators to be approved for clinical use in Europe. The FDC of a LAMA, umeclidinium (UMEC), and a LABA, vilanterol (VI) (62.5/25 μg), known as UMEC/VI (Anoro, GlaxoSmithKline), was approved by the FDA in December 2013, by the EMA in May, 2014 and in Japan in July, 2014. In a recent study, the use of once-daily UMEC/VI FDC over 24 weeks resulted in significant improvements of trough FEV1 when compared to placebo as well as monotherapy of the individual components (Citation54).

Table 2.  LABA/LAMA fixed-dose combinations in late-stage clinical development

In Phase II studies, QVA149 showed a fast onset of action with a statistically significant (p < 0.0001) increase in FEV1 over placebo, indacaterol (300 μg and 600 μg) 5 minutes post-dose on day 1 and a significant (p < 0.05) increase in FEV1 over placebo, indacaterol (300 μg and 600 μg) at all time-points on day 1 (Citation55). In a cardiovascular safety-orientated trial, QVA149 was well tolerated with an acceptable AE profile (Citation56).

QVA149 is being investigated in a large ongoing, Phase III programme called IGNITE comprising 11 studies of more than 10,000 patients. One trial (ILLUMINATE) compared QVA149 with SFC in 523 patients with no history of exacerbations within the preceding year in a double-blind, double-dummy, parallel-group design (Citation40). QVA149 provided significantly better, clinically relevant improvements in lung function with a rapid onset of action; at 26 weeks, FEV1 AUC0–12h was significantly higher with QVA149, with a treatment difference of 0·138 L (p < 0·0001 versus SFC) (see Figure ). Significant improvements in TDI scores, with a treatment difference of 0.76 (p = 0.003 versus SFC), and reduced use of rescue medication was also seen with QVA149. The study design allowed insights into the impact of ICS withdrawal—at Week 26 patients who had been on ICS therapy at enrollment showed similarly higher values for FEV1 with QVA149 compared with SFC to those who had not been on ICS treatment. This study supports the approach of using one or more bronchodilators for symptomatic COPD at low risk of exacerbation without ICS.

Figure 1. Comparison of FEV1 for QVA149 versus fluticasone/salmeterol. The ILLUMINATE double-blind, randomised trial of 523 patients. The primary study endpoint was FEV1 AUC0–12 h at 26 weeks for QVA149 vs. salmeterol/fluticasone (Citation40). At Week 26, FEV1 AUC0–12 h was significantly higher with QVA149 compared with fluticasone/salmeterol combination, with a significant and clinically meaningful treatment difference of 138 mL (95% CI 100–176; p < 0.0001).

Figure 1. Comparison of FEV1 for QVA149 versus fluticasone/salmeterol. The ILLUMINATE double-blind, randomised trial of 523 patients. The primary study endpoint was FEV1 AUC0–12 h at 26 weeks for QVA149 vs. salmeterol/fluticasone (Citation40). At Week 26, FEV1 AUC0–12 h was significantly higher with QVA149 compared with fluticasone/salmeterol combination, with a significant and clinically meaningful treatment difference of 138 mL (95% CI 100–176; p < 0.0001).

Conclusion

The role of ICS in the treatment paradigm for COPD is changing. This is driven by the growing evidence of increased risk of pneumonia, and a new class of LABA/LAMA FDC which simplifies dual bronchodilation, presenting a plausible alternative for some patients. As the evidence base for dual bronchodilation expands, it is likely that maximal bronchodilation will move up the treatment algorithm, with ICS reserved for those with more severe disease who are not controlled on dual therapy or where the clinical phenotype is known to receive therapeutic benefit. This change has already manifested in some COPD algorithms, such as those at Tayside, representing a significant change in recommended prescribing practice. Co-prescribing of LABA and LAMA is utilised much less frequently than LAMA monotherapy, ICS/LABA combinations, and the triple combination of LAMA plus ICS/LABA in recent reviews of prescribing practice (Citation12).

The key issue is the balance of risk-benefit of ICS in individual patients; the risk of pneumonia versus the benefit of reduced exacerbations. With the availability of new FDC, dual bronchodilation can be prescribed without increasing the dose of the LABA or LAMA component, and delivered in a simple, once-daily dosing regimen as a single inhaler. The once-daily dosing may be quite important for COPD patients, with evidence suggesting that compliance and adherence to treatment plans is historically poor, with COPD patients particularly vulnerable to treatment adherence lapses (Citation57). However, further data of the equivalence of LABA/LAMA with ICS/LABA in respect to exacerbation frequency is required.

For those patients already receiving ICS, the key concern is the risk of precipitating an exacerbation by ICS cessation, therefore careful review and individualised patient management is required. A patient's response to treatment, their preference, the likely side effects, the risk of exacerbation and financial cost implications require consideration. However for those patients with FEV1 > 50% without frequent exacerbations, it is likely that the risk-benefit is in favour of considering stopping ICS treatment. ICS withdrawal may be associated with worsening of COPD control so must be done carefully (Citation58). For those patients with the most advanced disease, triple therapy will remain the preferred treatment. The optimal posology of triple drug treatment requires further consideration, with the likely scenario of LABA/LAMA FDC co-prescribed with an ICS in a single inhaler emerging as the preferred option.

Declaration of Interests Statement

Aidan McManus, PhD CMPP and Ana Martins-Kaczor, PhD assisted Drs. Wilkie and Schembri to write a first draft of the manuscript based on a transcript and PowerPoint presentation given by Dr. Schembri, and provided subsequent editorial support as directed by Dr. Schembri. This editorial assistance was funded by Novartis UK, who reviewed the manuscript for medical accuracy but not content. These individuals did not meet the ICMJE criteria for authorship (www.icmje.org/).

Dr. Stuart Schembri has received honoraria and funding from GlaxoSmithKline, Astra-Zeneca, Boehringer Ingelheim, Novartis, Napp and Chiesi. He has attended advisory boards for Bayer, Chiesi and Novartis. Dr. Schembri is conducting clinical research with Novartis and Nycomed. Dr. Morven Wilkie has accepting funding from Pfizer in order to attend meetings. Dr. Simon Finch declares no conflict of interest.

Authors’ Contributions

Dr. Stuart Schembri conceived the review and its content, including all interpretation of the studies described, based on a presentation given at a COPD meeting in Scotland in June, 2013. Dr. Morven Wilkie and Dr. Simon Finch reviewed and commented on the first draft and approved the final version.

References

  • GOLD: Global strategy for the diagnosis, management, and prevention of COPD. http://www.goldcopd.org/uploads/users/files/GOLD_Report_2013_Feb20.pdf (accessed 30 October, 2013) 2013.
  • Yang IA, Clarke MS, Sim EH, Fong KM. Inhaled corticosteroids for stable chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2012; 7:CD002991.
  • Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Yates JC, Vestbo J, Investigators T. Salmeterol and fluticasone propionate and survival in chronic obstructive pulmonary disease. N Engl J Med 2007; 356:775–789.
  • National Clinical Guideline Centre. (2010) Chronic obstructive pulmonary disease: management of chronic obstructive pulmonary disease in adults in primary and secondary care. London: National Clinical Guideline Centre. Available from: http://guidance.nice.org.uk/CG101/Guidance/pdf/English Accessed February 2015.
  • Vestbo J, Sorensen T, Lange P, Brix A, Torre P, Viskum K. Long-term effect of inhaled budesonide in mild and moderate chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 1997; 353:1819-1823.
  • Mehuys E, Boussery K, Adriaens E, Van Bortel L, De Bolle L, Van Tongelen I, Remon JP, Brusselle G. COPD management in primary care: an observational, community pharmacy-based study. Ann Pharmacother 2010; 44:257–266.
  • Price D, Yawn B, Brusselle G, Rossi A. Risk-to-benefit ratio of inhaled corticosteroids in patients with COPD. Prim Care Respir J 2013; 22:92–100.
  • Janson C, Larsson K, Lisspers KH, Ställberg B, Stratelis G, Goike H, Jörgensen L, Johansson G. Pneumonia and pneumonia related mortality in patients with COPD treated with fixed combinations of inhaled corticosteroid and long acting β2 agonist: observational matched cohort study (PATHOS). BMJ 2013; 346:f3306.
  • Sharafkhaneh A, Southard JG, Goldman M, Uryniak T, Martin UJ. Effect of budesonide/formoterol pMDI on COPD exacerbations: a double-blind, randomized study. Respir Med 2012; 106:257–268.
  • Dransfield MT, Bourbeau J, Jones PW, Hanania NA, Mahler DA, Vestbo J, Wachtel A, Martinez FJ, Barnhart F, Sanford L, et al. Once-daily inhaled fluticasone furoate and vilanterol versus vilanterol only for prevention of exacerbations of COPD: two replicate double-blind, parallel-group, randomised controlled trials. Lancet Respir Med 2013; 1:210–223.
  • Bousquet J. Inhaled corticosteroids in severe COPD. Lancet Respiratory Medicine 2013; 1(3):177–178.
  • Tashkin DP, Ferguson GT. Combination bronchodilator therapy in the management of chronic obstructive pulmonary disease. Respir Res 2013; 14:49.
  • Bateman ED, Ferguson GT, Barnes N, Gallagher N, Green Y, Henley M, Banerji D. Dual bronchodilation with QVA149 versus single bronchodilator therapy: the SHINE study. Eur Respir J 2013; 42(6):1441–1445.
  • Connolly CK, Alcock SM, Prescott RJ. Management and control of asthma as assessed by actual/best function and corticosteroid use 1980-1993/4. Eur Respir J 1998; 12:859–864.
  • Bourbeau J, Rouleau MY, Boucher S. Randomised controlled trial of inhaled corticosteroids in patients with chronic obstructive pulmonary disease. Thorax 1998; 53:477–482.
  • Renkema TE, Schouten JP, Koeter GH, Postma DS. Effects of long-term treatment with corticosteroids in COPD. Chest 1996; 109:1156–1162.
  • Weir DC, Bale GA, Bright P, Sherwood Burge P. A double-blind placebo-controlled study of the effect of inhaled beclomethasone dipropionate for 2 years in patients with nonasthmatic chronic obstructive pulmonary disease. Clin Exp Allergy 1999; 29 Suppl 2:125–128.
  • Senderovitz T, Vestbo J, Frandsen J, Maltbaek N, Norgaard M, Nielsen C, Kampmann JP. Steroid reversibility test followed by inhaled budesonide or placebo in outpatients with stable chronic obstructive pulmonary disease. Danish Soc Respir Med Respir Med 1999; 93:715–718.
  • Paggiaro PL, Dahle R, Bakran I, Frith L, Hollingworth K, Efthimiou J. Multicentre randomised placebo-controlled trial of inhaled fluticasone propionate in patients with chronic obstructive pulmonary disease. International COPD Study Group. Lancet 1998; 351:773–780.
  • Burge PS, Calverley PM, Jones PW, Spencer S, Anderson JA, Maslen TK. Randomised, double blind, placebo controlled study of fluticasone propionate in patients with moderate to severe chronic obstructive pulmonary disease: the ISOLDE trial. BMJ 2000; 320:1297–1303.
  • Calverley P, Pauwels R, Vestbo J, Jones P, Pride N, Gulsvik A, Anderson J, Maden C. STeroids TRoI, long-acting beta2 agonists study g: Combined salmeterol and fluticasone in the treatment of chronic obstructive pulmonary disease: a randomised controlled trial. Lancet 2003; 361:449–456.
  • Kardos P, Wencker M, Glaab T, Vogelmeier C. Impact of salmeterol/fluticasone propionate versus salmeterol on exacerbations in severe chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2007; 175: 144–149.
  • Lapperre TS, Snoeck-Stroband JB, Gosman MM, Jansen DF, van Schadewijk A, Thiadens HA, Vonk JM, Boezen HM, Ten Hacken NH, Sont JK, Rabe KF, Kerstjens HA, Hiemstra PS, Timens W, Postma DS, Sterk PJ; Groningen Leiden Universities: Corticosteroids in Obstructive Lung Disease Study Group. Effect of fluticasone with and without salmeterol on pulmonary outcomes in chronic obstructive pulmonary disease: a randomized trial. Ann Intern Med 2009; 151:517–527.
  • Bafadhel M, McKenna S, Terry S, Mistry V, Reid C, Haldar P, McCormick M, Haldar K, Kebadze T, Duvoix A, Lindblad K, Patel H, Rugman P, Dodson P, Jenkins M, Saunders M, Newbold P, Green RH, Venge P, Lomas DA, Barer MR, Johnston SL, Pavord ID, Brightling CE. Acute exacerbations of chronic obstructive pulmonary disease: identification of biologic clusters and their biomarkers. Am J Respir Crit Care Med 2011; 184:662–671.
  • Siva R, Green RH, Brightling CE, Shelley M, Hargadon B, McKenna S, Monteiro W, Berry M, Parker D, Wardlaw AJ, Pavord ID. Eosinophilic airway inflammation and exacerbations of COPD: a randomised controlled trial. Eur Respir J 2007; 29:906–913.
  • Nannini LJ, Poole P, Milan SJ, Kesterton A. Combined corticosteroid and long-acting beta2-agonist in one inhaler versus inhaled corticosteroids alone for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2013; 8:CD006826.
  • Celli BR, Thomas NE, Anderson JA, Ferguson GT, Jenkins CR, Jones PW, Vestbo J, Knobil K, Yates JC, Calverley PM. Effect of pharmacotherapy on rate of decline of lung function in chronic obstructive pulmonary disease: results from the TORCH study. Am J Respir Crit Care Med 2008; 178:332–338.
  • Postma DS, Calverley P. Inhaled corticosteroids in COPD: a case in favour. Eur Respir J 2009; 34:10–12.
  • Suissa S, Barnes PJ. Inhaled corticosteroids in COPD: the case against. Eur Respir J 2009; 34:13–16.
  • Singh SD, Whale C, Houghton N, Daley-Yates P, Kirby SM, Woodcock AA. Pharmacokinetics and systemic effects of inhaled fluticasone propionate in chronic obstructive pulmonary disease. Br J Clin Pharmacol 2003; 55:375–381.
  • Miller DP, Watkins SE, Sampson T, Davis KJ. Long-term use of fluticasone propionate/salmeterol fixed-dose combination and incidence of cataracts and glaucoma among chronic obstructive pulmonary disease patients in the UK General Practice Research Database. Int J Chron Obstruct Pulmon Dis 2011; 6:467–476.
  • Flynn RW, MacDonald TM, Hapca A, MacKenzie IS, Schembri S. Quantifying the real life risk profile of inhaled corticosteroids in COPD by record linkage analysis. Respir Res. 2014 Nov 19;15:141.
  • Duerden M. Prevention of death in COPD. N Engl J Med 2007; 356:2212; author reply 2213-2214.
  • Wedzicha JA, Calverley PM, Seemungal TA, Hagan G, Ansari Z, Stockley RA. Investigators I: The prevention of chronic obstructive pulmonary disease exacerbations by salmeterol/fluticasone propionate or tiotropium bromide. Am J Respir Crit Care Med 2008; 177:19–26.
  • Ernst P, Gonzalez AV, Brassard P, Suissa S. Inhaled corticosteroid use in chronic obstructive pulmonary disease and the risk of hospitalization for pneumonia. Am J Respir Crit Care Med 2007; 176:162–166.
  • Loke YK, Cavallazzi R, Singh S. Risk of fractures with inhaled corticosteroids in COPD: systematic review and meta-analysis of randomised controlled trials and observational studies. Thorax 2011; 66:699–708.
  • Suissa S, Kezouh A, Ernst P. Inhaled corticosteroids and the risks of diabetes onset and progression. Am J Med 2010; 123:1001–1006.
  • Caughey GE, Preiss AK, Vitry AI, Gilbert AL, Roughead EE. Comorbid diabetes and COPD: impact of corticosteroid use on diabetes complications. Diabetes Care 2013; 36:3009–3014.
  • Crim C, Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, Jones PW, Willits LR, Yates JC, Vestbo J. Pneumonia risk in COPD patients receiving inhaled corticosteroids alone or in combination: TORCH study results. Eur Respir J 2009; 34:641–647.
  • Vogelmeier CF, Bateman ED, Pallante J, Alagappan VKT, D’Andrea P, Chen H, Banerji D. Efficacy and safety of once-daily QVA149 compared with twice-daily salmeterol–fluticasone in patients with chronic obstructive pulmonary disease (ILLUMINATE): a randomised, double-blind, parallel group study. Lancet Respir Med 2013; 1:51–60.
  • Singanayagam A, Chalmers JD, Akram AR, Hill AT. Impact of inhaled corticosteroid use on outcome in COPD patients admitted with pneumonia. Eur Respir J 2011; 38:36–41.
  • Torres A, Ewig S. The strange case of community-acquired pneumonia in COPD. Chest 2011; 139:483–485.
  • Doherty DE, Tashkin DP, Kerwin E, Knorr BA, Shekar T, Banerjee S, Staudinger H. Effects of mometasone furoate/formoterol fumarate fixed-dose combination formulation on chronic obstructive pulmonary disease (COPD): results from a 52-week Phase III trial in subjects with moderate-to-very severe COPD. Int J Chron Obstruct Pulmon Dis 2012; 7:57–71.
  • Corrado A, Rossi A. How far is real life from COPD therapy guidelines? An Italian observational study. Respir Med 2012; 106:989–997.
  • Jebrak G, Initiatives B. (COPD routine management in France: are guidelines used in clinical practice?). Rev Mal Respir 2010; 27:11–18.
  • Sheng X, Murphy MJ, MacDonald TM, Schembri S, Simpson W, Winter J, Winter JH, Wei L. Effect of statins on total cholesterol concentrations, cardiovascular morbidity, and all-cause mortality in chronic obstructive pulmonary disease: a population-based cohort study. Clin Ther 2012; 34:374–384.
  • Short PM, Lipworth SI, Elder DH, Schembri S, Lipworth BJ. Effect of beta blockers in treatment of chronic obstructive pulmonary disease: a retrospective cohort study. BMJ 2011; 342:d2549.
  • Consortium SM. Salmeterol/fluticasone 50/500 micrograms inhaler (Seretide 500 Accuhaler). NHS Scotland, 2008.
  • Cazzola M, Molimard M. The scientific rationale for combining long-acting beta2-agonists and muscarinic antagonists in COPD. Pulm Pharmacol Ther 2010; 23:257–267.
  • van Noord JA, Aumann JL, Janssens E, Smeets JJ, Zaagsma J, Mueller A, Cornelissen PJ. Combining tiotropium and salmeterol in COPD: Effects on airflow obstruction and symptoms. Respir Med 2010; 104:995–1004.
  • van der Molen T, Cazzola M. Beyond lung function in COPD management: effectiveness of LABA/LAMA combination therapy on patient-centred outcomes. Prim Care Respir J 2012; 21:101–108.
  • Mahler DA, D’Urzo A, Bateman ED, Ozkan SA, White T, Peckitt C, Lassen C, Kramer B. Intrust, investigators I-s: Concurrent use of indacaterol plus tiotropium in patients with COPD provides superior bronchodilation compared with tiotropium alone: a randomised, double-blind comparison. Thorax 2012; 67:781–788.
  • Wang J, Jin D, Zuo P, Wang T, Xu Y, Xiong W. Comparison of tiotropium plus formoterol to tiotropium alone in stable chronic obstructive pulmonary disease: a meta-analysis. Respirology 2011; 16:350–358.
  • Donohue JF, Maleki-Yazdi MR, Kilbride S, Mehta R, Kalberg C, Church A. Efficacy and safety of once-daily umeclidinium/vilanterol 62.5/25 mcg in COPD. Respiratory Medicine 2013; 107:1538–1546.
  • van Noord JA, Buhl R, Laforce C, Martin C, Jones F, Dolker M, Overend T. QVA149 demonstrates superior bronchodilation compared with indacaterol or placebo in patients with chronic obstructive pulmonary disease. Thorax 2010; 65:1086–1091.
  • Van de Maele B, Fabbri LM, Martin C, Horton R, Dolker M, Overend T. Cardiovascular safety of QVA149, a combination of Indacaterol and NVA237, in COPD patients. COPD 2010; 7:418–427.
  • Bourbeau J, Bartlett SJ. Patient adherence in COPD. Thorax 2008; 63:831–838.
  • Nadeem NJ, Taylor SJ, Eldridge SM. Withdrawal of inhaled corticosteroids in individuals with COPD—a systematic review and comment on trial methodology. Respir Res 2011; 12:107.