ArticlePDF AvailableLiterature Review

Inhaled Combination Therapy With Long-Acting 2-Agonists and Corticosteroids in Stable COPD

Authors:

Abstract and Figures

Long-acting beta(2)-agonists (LABAs) have been shown to be effective first-line bronchodilators in the treatment of COPD patients, and inhaled corticosteroids (ICSs) have been shown to reduce the frequency and/or severity of exacerbations in COPD patients. The concomitant use of a LABA and an ICS can influence both airway obstruction (ie, smooth muscle contraction, increased cholinergic tone, and loss of elastic recoil), and airway inflammation (ie, increased numbers of neutrophils, macrophages, and CD8+ lymphocytes, elevated interleukin-8 and tumor necrosis factor-alpha levels, and protease/antiprotease imbalance). They are also able to reduce the total number of bacteria adhering to the respiratory mucosa in a concentration-dependent manner without altering the bacterial tropism for mucosa, and to preserve ciliated cells. Several clinical trials support the concept of inhaled combination therapy with LABAs and corticosteroids in stable COPD patients. This type of therapy not only improves airflow obstruction but also provides clinical benefits, as manifested by sustained reduction in overall symptoms, improvements in health-related quality of life, and reductions in exacerbations. All of these effects are very important because, despite recent advances in our understanding of COPD and its treatment, therapy remains suboptimal for a considerable number of patients.
Content may be subject to copyright.
Inhaled Combination Therapy With
Long-Acting
2
-Agonists and
Corticosteroids in Stable COPD*
Mario Cazzola, MD, FCCP; and Ronald Dahl, MD
Long-acting
2
-agonists (LABAs) have been shown to be effective first-line bronchodilators in the
treatment of COPD patients, and inhaled corticosteroids (ICSs) have been shown to reduce the
frequency and/or severity of exacerbations in COPD patients. The concomitant use of a LABA and
an ICS can influence both airway obstruction (ie, smooth muscle contraction, increased cholin-
ergic tone, and loss of elastic recoil), and airway inflammation (ie, increased numbers of
neutrophils, macrophages, and CD8 lymphocytes, elevated interleukin-8 and tumor necrosis
factor- levels, and protease/antiprotease imbalance). They are also able to reduce the total
number of bacteria adhering to the respiratory mucosa in a concentration-dependent manner
without altering the bacterial tropism for mucosa, and to preserve ciliated cells. Several clinical
trials support the concept of inhaled combination therapy with LABAs and corticosteroids in
stable COPD patients. This type of therapy not only improves airflow obstruction but also
provides clinical benefits, as manifested by sustained reduction in overall symptoms, improve-
ments in health-related quality of life, and reductions in exacerbations. All of these effects are
very important because, despite recent advances in our understanding of COPD and its
treatment, therapy remains suboptimal for a considerable number of patients.
(CHEST 2004; 126:220 –237)
Key words: inhaled corticosteroids; long-acting
2
-agonists
Abbreviations: cAMP cyclic adenosine 35-monophosphate; C/EBP CCAAT-enhancer binding protein;
EUROSCOP European Respiratory Society Study on COPD; GR glucocorticoid receptor; ICS inhaled cortico-
steroid; IL interleukin; LABA long-acting
2
-agonist; NF-B nuclear factor-B; PEF peak expiratory flow;
QOL quality of life; SABA short-acting
2
-agonist; SEK Swedish kronor; SGRQ St. George Respiratory
Questionnaire; TNF tumor necrosis factor; TRISTAN Trial of Inhaled Steroids and Long-Acting
2
-Agonists
C
OPD is a multicomponent disease that includes
airway inflammation, airflow limitation, muco-
ciliary dysfunction, and airway structural changes
1
(Fig 1). In particular, obstruction is associated with
an airway inflammatory profile consisting mainly of
an increased number of T lymphocytes (predomi-
nantly CD8 cells), macrophages, and neutro-
phils.
2,3
Airway inflammation also involves inflamma-
tory mediators such as leukotriene B4, interleukin
(IL)-8 and tumor necrosis factor (TNF)-,
4
which
are generally considered to be important mediators
in neutrophil recruitment. Oxidative stress and an
imbalance between proteases and antiproteases in
the lung also have been implicated in the pathophys-
iology of the condition.
4
All COPD components contribute to a complex of
lung function changes, symptoms, and exacerbations,
which affect the health status and, ultimately, the
survival of the patient.
1
It is obvious, therefore, that
a correct therapeutic approach must try to interfere
with these components. The recent evidence-based
Global Initiative for Chronic Obstructive Lung Dis-
ease guidelines
1
recommend that the overall ap-
proach to managing stable COPD patients should be
characterized by a stepwise increase in treatment,
*From the Department of Respiratory Medicine (Dr. Cazzola),
Unit of Pneumology and Allergology, Antonio Cardarelli Hospi-
tal, Naples, Italy; and the Department of Respiratory Diseases
(Dr. Dahl), University Hospital Aarhus, Aarhus, Denmark.
Dr. Cazzola has received fees for participating to Advisory Boards
from Altana, AstraZeneca, and Novartis, for consulting from
Chiesi Farmaceutici and GSK, and for speaking from AstraZen-
eca, Boehringer Ingelheim, GSK, Menarini Farmaceutici, Novar-
tis, and Pfizer, and research-related grants from GSK. Dr. Dahl
has received fees for participating in Advisory Boards from
Altana, Boehringer-Ingelheim, and GSK, for speaking from
Schering-Plough, MSD, and ALK-Abello, and research-related
grants from Astra-Zeneca, GSK, ALK-Abello, Pfizer, Boehringer-
Ingelheim, Novartis, Almirall, MSD, and UCB.
Manuscript received June 25, 2003; revision accepted October
14, 2003.
Reproduction of this article is prohibited without written permis-
sion from the American College of Chest Physicians (e-mail:
permissions@chestnet.org).
Correspondence to: Mario Cazzola, MD, FCCP, Via del Parco
Margherita 24, 80121 Napoli, Italy; e-mail mcazzola@qubisoft.it
220 Reviews
depending on the severity of the disease. To date, no
medication has been shown to alter the rate of
decline of FEV
1
, but bronchodilator medications are
central to symptom management in COPD.
5
Among
the currently available bronchodilators, long-acting
inhaled agents are the most convenient.
6
However,
many patients remain symptomatic despite the opti-
mal use of these bronchodilators, and, consequently,
it can be necessary to add other drugs to the
therapeutic regimen. A body of evidence indicates
that combining different bronchodilators may im-
prove efficacy and decrease the risk of side effects
compared to increasing the dose of a single agent.
1,5
The Global Initiative for Chronic Obstructive
Lung Disease guidelines
1
are more cautious in sug-
gesting the use of corticosteroids. They state that
regular treatment with inhaled corticosteroids (ICSs)
should be prescribed only for symptomatic COPD
patients with a documented spirometric response to
corticosteroids or in those with an FEV
1
50% of
predicted values, and repeated exacerbations requir-
ing treatment with antibiotics and/or oral corticoste-
roids.
Use of Long-Acting
2
-Agonists in COPD
Long-acting
2
-agonists (LABAs) have been
shown to be elective first-line bronchodilators in the
treatment of patients with COPD.
5,6
They safely
attenuate airflow obstruction, decrease the fre-
quency and severity of symptoms by reducing the
amount of dynamic hyperinflation, and improve
quality of life (QOL). Interestingly, several studies
have documented that the LABAs formoterol
7,8
and
salmeterol
9,10
are more effective than ipratropium
bromide for the regular treatment of COPD. This
finding is important because for many years ipratro-
pium has been considered to be the first-line therapy
for long-term treatment of stable patients who are
symptomatic with COPD.
11
LABAs can usefully be prescribed for the com-
bined bronchodilator treatment of COPD.
12,13
In
particular, the addition of a LABA to an anticholin-
ergic compound appears to be more efficacious than
adding a short-acting
2
-agonist (SABA) in stable
patients with COPD.
14
In addition to prolonged bronchodilatation,
LABAs exert other additive effects that may be of
clinical relevance in COPD
15
(Table 1). For exam-
ple, data indicate that LABAs can increase the level
of cyclic adenosine 35-monophosphate (cAMP) in
neutrophils, thereby inhibiting neutrophil adhesion,
accumulation, and activation, and inducing apopto-
sis.
15
In particular, salmeterol inhibits neutrophil
adhesion to bronchial epithelial cells.
16
Formoterol
inhibits chemotaxis to platelet-activating factor
17
over the concentration range 10
7
to 10
4
mol/L.
Neutrophil activation also was attenuated, as evi-
denced by reductions in the release of superoxide,
18
IL-8,
19
and bacterial permeability-increasing pro-
tein,
20
which are not associated with the SABA
albuterol. However, in general, these effects were
only significant at relatively high concentrations (ie,
10
6
mol/L) and were not reversed by proprano
-
lol, suggesting that they were not mediated by
2
-adrenoceptors. The end result is a possible reduc
-
tion in the number and activation status of neutro-
phils in airway tissue and in the airway lumen
21
Figure 1. Pathophysiologic/clinical features of COPD. The multiple components of COPD combine
to produce airflow limitation, which is a key characteristic of the disease.
Table 1—LABA Mechanisms of Action in COPD
Bronchodilation
Reduced lung hyperinflation
Increased mucociliary transport
Mucosal cytoprotection
Antineutrophil activity
www.chestjournal.org CHEST / 126/1/JULY, 2004 221
(Table 2). LABAs also may decrease the number of
neutrophils that adhere to the vascular endothelium
at sites of inflammation and may reduce the amount
of plasma leakage,
22
but the relevance of these
findings to COPD patients is unclear. It is notewor-
thy that glucocorticoids have been shown to increase
high-affinity -agonist binding in human neutro-
phils.
23
It is also possible that LABAs may increase
the peripheral deposition of ICSs, thus enhancing
their antiinflammatory activity.
24
Use of Corticosteroids in COPD
The role of corticosteroids in the management of
COPD remains controversial. ICSs are commonly
prescribed in high doses on the basis that COPD is
like poorly responsive asthma, but evidence that they
are beneficial in patients with pure COPD is still
weak. In effect, although it is well-known that corti-
costeroids are effective at suppressing airway inflam-
mation, their effect on the inflammation in COPD is
still unclear.
Impact of Corticosteroids on Inflammation in
COPD
Some studies
25–28
have indicated that these agents
apparently do not exert a major influence on inflam-
matory cells and mediators in the sputum of stable
patients with COPD, but other reports
29–36
have
suggested that treatment with corticosteroids may
induce biological responses that may be associated
with changes in some clinical outcomes and endo-
scopic findings in these patients (Table 3).
In vitro corticosteroids attenuate neutrophil re-
cruitment and activation,
29
and reduce neutrophil
chemotaxis.
30
In vivo, they attenuate sputum chemo-
tactic activity, increase neutrophil elastase inhibitory
capacity,
31
and induce a decrease in both neutrophils
and total cells in induced sputum.
32,33
Moreover,
they reduce the numbers of mucosal mast cells
34
and
subepithelial mast cells, and reduce the epithelial
CD8/CD4 ratio,
35
although they have no effect on
the major inflammatory cell types in COPD. ICSs
also induce significant reductions in the BAL fluid
levels of IL-8 and myeloperoxidase, as well as reduc-
tions in cell numbers, the proportion of neutrophils,
symptom score, and bronchitis index.
36
These findings seem to support the use of ICSs in
treating COPD, although long-term treatment with
ICSs must be associated with a high risk of adverse
systemic effects and involves unnecessary expense.
37
Unfortunately, however, clinical trials on the effects
of corticosteroids in COPD have reported conflicting
results. Consequently, there is considerable contro-
versy concerning the utility of these agents for the
long-term treatment of patients with COPD.
37,38
Clinical Effects of Corticosteroids in COPD
Negative Findings: Some trials
39–41
have seemed
to indicate that only those patients with an asthmatic
component to their disease appear to benefit from
corticosteroids. Thus, in the study by Chanez and
colleagues,
39
12 of 25 unselected patients in whom
COPD had been clinically diagnosed responded to a
daily oral dose of prednisolone, 1.5 mg/kg body
weight, for 15 days, with an increase in FEV
1
of at
least 12% from the baseline value and an absolute
value of 200 mL, measured at the end of treatment.
By comparison with nonresponders, responders had
a significantly larger number of eosinophils and
higher levels of eosinophil cationic protein in their
BAL fluid, and the responders also had a thicker
reticular basement membrane than the nonre-
sponders. Pizzichini and colleagues
40
reported that
an improvement in QOL score and FEV
1
after a
short-term course of prednisone therapy in smokers
with chronic obstructive bronchitis was paralleled by
a significant reduction in eosinophilia and eosinophil
activation, as indicated by sputum eosinophil cationic
protein levels, but not by changes in neutrophils or
neutrophil proteases. Patients without sputum eosin-
ophilia did not show clinical benefit from short-term
prednisone therapy. Nishimura and colleagues,
41
in a
trial of patients with COPD, found a significant
improvement in FEV
1
in a minority of patients (5 of
30 patients) receiving 3,000 g per day beclometha-
sone dipropionate over a 4-week treatment period.
However, some of these responders showed a posi-
tive response to the bronchodilator challenge, as well
as an elevated serum IgE level or eosinophil count,
which is suggestive of the presence of an asthmatic
component to their airflow obstruction.
Besides, four large studies (ie, the European
Respiratory Society Study on COPD [EURO-
Table 2Antineutrophil Effects of Salmeterol
21
Variables Placebo
Salmeterol,
50 g twice
daily 6wk
Neutrophil numbers
BAL fluid, % 2.1 1.6*
Bronchial biopsy, cells/mm
2
21.4 14.3†
Neutrophil mediator release
BAL IL-8, pg/mL 73.0 61.0*
BAL lipocalin, mg/L 14.1 10.3‡
Serum myeloperoxidase, mg/L 302 249*
*p 0.05.
†p 0.04.
‡p 0.03.
222 Reviews
SCOP]
42
; the Copenhagen City Lung Study
43
; the
Inhaled Steroids in Obstructive Lung Disease trial
44
;
and the American Lung Health-2
45
) have demon-
strated that these agents did not have any worthwhile
effect on the rate of decline in FEV
1
when asthma
was rigorously excluded (Table 4), indicating that
there is no effect of ICSs on the progressive inflam-
matory disease process.
37
In effect, there are three principal arguments
against the use of ICSs in COPD, as follows: neu-
trophilic inflammation (which is characteristic in
COPD) is generally resistant to corticosteroids; cor-
ticosteroids prolong the survival of neutrophils by
inhibiting apoptosis; and corticosteroid therapy fails
to suppress cytokines such as TNF- and IL-8,
which are elevated in patients with COPD.
37
More-
Table 3Impact of Corticosteroids on Inflammation in COPD*
Study Type of Patients Enrolled
Treatment, Study
Duration Outcomes
Negative studies
Thompson et al
25
Current smokers with chronic
bronchitis and at least mild
obstruction
Beclomethasone, 1 g/d for
6 weeks
Small increase in FEV
1
, small decrease in macroscopic
bronchioscopic index of bronchial inflammation, no
reduction in the number of neutrophils in BALF
Keatings et al
26
Patients with severe COPD
(mean FEV
1
, 35%
predicted)
Budesonide, 800 g twice
daily for 2 wk
No clinical benefit in either lung function or symptom
scores, no significant change in the inflammatory
indices as measured by total and differential cell
counts and concentrations of TNF-, eosinophil
activation markers, eosinophilic cationic protein, and
eosinophil peroxidase, and neutrophil activation
markers myeloperoxidase and human neutrophil
lipocalin
Oral prednisolone, 30 mg
daily for 2 weeks
Sputum eosinophil number, eosinophilic cationic
protein, and eosinophil peroxidase not modified
Culpitt et al
27
Patients with stable COPD Fluticasone, 500 g twice
daily for 4 wk
No clinical benefit in terms of lung function or
symptom scores, no change in induced sputum
inflammatory cells, percentage of neutrophils, IL-8
levels, supernatant elastase activity, MMP-1, MMP-
9, and the antiproteases secretory leukoprotease
inhibitor and tissue inhibitor of MMP-1 levels
Loppow et al
28
Patients with chronic
bronchitis (mean FEV
1
,
83.4% predicted)
Fluticasone, 500 g twice
daily for 4 wk
No improvement in lung function or inflammatory
parameters, such as the concentration of exhaled
nitric oxide, differential cell counts in induced
sputum, and the number of cells positive for
inducible nitric oxide synthase, as well as the levels
of lactate dehydrogenase, eosinophilic cationic
protein, neutrophil elastase and IL-8 in sputum
supernatants
Positive studies
Llewellyn-Jones et al
31
Patients with clinically stable,
smoking-related chronic
bronchitis and emphysema
(mean FEV
1
, 0.71 L)
Fluticasone, 1.5 mg/d for
8wk
No effect on peripheral neutrophils or on sputum
albumin and myeloperoxidase concentrations, but
reduction in the neutrophil chemotactic activity of
sputum and beneficial effect on the
proteinase/antiproteinase balance
Confalonieri et al
32
Patients with stable COPD
(mean FEV
1
, 60.2%
predicted)
Beclomethasone, 500 g
three times daily for 2
mo
Reduction in both neutrophils and total cells in
induced sputum, no change in spirometry and blood
gases
Yildiz et al
33
Clinically stable COPD
patients
Fluticasone, 1,500 g/d
for2mo
No significant changes in the number of peripheral
blood neutrophils, blood gases, and spirometry, but
decrease in the total cell number and the number of
neutrophils in induced sputum
Gizycki et al
34
Patients with mild-to-severe
COPD (FEV
1
,2580%
predicted)
Fluticasone, 500 g twice
daily for 3 mo
Significant decrease in the numbers of mucosal mast
cells, improvement in symptoms
Hattotuwa et al
35
Patients with mild-to-severe
stable COPD (mean FEV
1
,
2580% predicted)
Fluticasone, 500 g twice
daily for 3 mo
No effect on the major inflammatory cell types in
COPD, but reduced epithelial CD8/CD4 ratio and
subepithelial mast cell number
Balbi et al
36
Stable COPD patients with
mild disease
Beclomethasone, 1.5 mg/d
for6wk
Reductions in the lavage levels of IL-8 and
myeloperoxidase, in cell numbers, neutrophil
proportion, symptom score, and bronchitis index
*BALF BAL fluid; MMP matrix metalloproteinase.
www.chestjournal.org CHEST / 126/1/JULY, 2004 223
over, a population-based cohort study
46
documented
that ICS use after hospitalization for COPD was not
found to reduce mortality and morbidity, but this
study had only 979 persons and could not estimate
the benefit with great precision.
Positive Findings: Notwithstanding these nega-
tive findings, evidence is accumulating to support the
therapeutic use of corticosteroids, at least in patients
with more advanced COPD.
47
For example, it was
reported that treatment with an ICS for 3 months
improved prebronchodilator airflow obstruction and
oxygenation while decreasing dyspnea in patients
with moderate-to-severe COPD.
48
Reductions in
exacerbation severity were seen in another study
49
of
patients with moderately severe disease who were
treated for 6 months with an inhaled glucocorticoid.
The Inhaled Steroids in Obstructive Lung Disease
study
44
reported that, in patients with moderate-to
severe-disease, 3 years of regular treatment with an
ICS resulted in fewer exacerbations, a reduced rate
of decline in health status, and higher FEV
1
values
than did placebo treatment. A recent systematic
review
50
of randomized placebo-controlled trials has
demonstrated a beneficial effect of ICSs in reducing
rates of COPD exacerbations. These agents also can
extend survival in patients with COPD.
51
Further-
more, the withdrawal of ICS therapy has been shown
to lead to deterioration in ventilatory function and to
increased exercise-induced dyspnea in patients with
severe irreversible airway obstruction.
52
All these findings lend support to the use of ICSs
in stable patients with COPD, and a convincing
demonstration of the effectiveness of these agents in
COPD patients has come from the COPE study (an
investigation of COPD in the Department of Pal-
monology, Enschede, the Netherlands).
53
This study
showed that the discontinuation of therapy with ICSs
was associated with a more rapid onset and a higher
risk of recurrence of exacerbations, and with a
significant deterioration in aspects of health-related
QOL, in the majority of patients.
53
However, 40% of
subjects in the COPE study experienced no unto-
ward effect from the withdrawal of ICSs.
53
This
finding indicates that there is an urgent need to
identify which subgroups of COPD patients respond
well to prolonged inhaled glucocorticoid therapy.
This is not an easy task, since COPD comprises
various diseases, which may differ in their pathologic,
clinical, and functional features, and in the biological
phenomena that cause and maintain airway inflamma-
tion. There also may be differences between the base-
line disease state (ie, in the absence of a current
exacerbation) and the disease state during an acute
exacerbation, and differences according to the degree
of airway obstruction or history of cigarette smoking.
Nevertheless, since very few therapies offer significant
benefits to patients with COPD, and since ICSs are
potentially beneficial in the treatment of this disease
(Table 5), the use of these agents remains a possible
therapeutic approach to stable patients with COPD.
Table 4Long-term Effect of ICSs in COPD*
Study
Patients Enrolled, No./Study
Duration Rate of FEV
1
Decline vs Placebo
Health Outcomes
EUROSCOP
42
1,277 patients with mild COPD
(mean FEV
1
, 77% predicted)/
F/U at 36 mo
No change with budesonide, 400 g
twice daily
Not evaluated
Copenhagen City Lung Study
43
290 patients with mild to
moderate COPD (mean FEV
1
86% of predicted value)/F/U
at 30 mo
No change with budesonide, 800 g
plus 400 g daily for 6 mo
followed by 400 g twice daily for
30 mo
No change in exacerbations
ISOLDE
44
750 patients, with moderate to
severe COPD (mean FEV
1
50% of predicted value)/F/U
at 36 mo
No change with fluticasone, 500 g
twice daily
Decreased exacerbations; reduced
rate in decline of the disease-
specific SGRQ
Health Lung Study-2
45
1,116 patients with mild-to-
moderate COPD (mean
FEV
1
, 64% predicted)/F/U at
40 mo
No change with triamcinolone, 600
g twice daily
Less airway reactivity; reduced
respiratory symptoms; slightly
reduced hospitalizations; loss of
bone mineral density; increased
skin bruising
*F/U follow up.
Table 5Clinical Efficacy of ICSs in COPD
Increased lung function
Decreased symptoms
Decreased No./severity of exacerbations
Decreased morbidity/mortality
Increased health status
224
Reviews
Therefore, until a test is developed that can distinguish
potential corticosteroid responders from nonre-
sponders, it is worthwhile giving all patients with
COPD a trial (3 to 6 months) of an ICS to determine
whether or not they respond.
54
Pharmacologic Rationale for Combining a
LABA and an ICS in COPD
It must always be emphasized that the use of ICSs
should not leave the use of a LABA out of consider-
ation. The concomitant use of a LABA and an ICS
can influence airway obstruction (eg, smooth muscle
contraction, increased cholinergic tone, and, per-
haps, bronchial hyperreactivity), mucociliary dys-
function (eg, reduced mucociliary transport and
mucosal damage), and airway inflammation (eg, in-
creased numbers of neutrophils, macrophages, and
CD8 lymphocytes, elevated levels of IL-8, TNF-,
and leukotrieneB4, protease/antiprotease imbalance,
and mucosal edema), as well as structural changes
(eg, glandular hypertrophy and goblet cell hyperpla-
sia) [Fig 2]. Furthermore, when a LABA is added to
an ICS, it has the potential for countering some of
the possible negative effects of the corticosteroid.
There is an exciting possibility that the observed
benefit from combining these two classes of drugs
might be due to a synergistic interaction, with the
resulting synergetic effect being greater than the
sum of responses achieved from each drug alone.
However, the basic molecular mechanism of such an
interaction is still to be fully identified (Fig 3).
Effect of Corticosteroids on LABAs
Corticosteroids can prevent, at least partially, ho-
mologous down-regulation of
2
-adrenoceptor num
-
bers and can induce an increase in the rate of
receptor synthesis through a process of extended
2
-adrenoceptor gene transcription.
55
Although air-
way smooth muscle is among the tissues least sus-
ceptible to homologous down-regulation, long-term
treatment with a LABA may result in tolerance to its
bronchodilator effects.
56
Thus, the effects of cortico-
steroids on
2
-adrenoceptor expression have the
potential for enhancing the airway relaxant response
to -adrenergic stimulation. An in vitro experimental
finding suggests that, at least in the rabbit, this effect
is correlated with increased -adrenoceptor expres-
sion in the tissue.
57
Moreover, the efficiency of
coupling between the
2
-adrenoceptor and Gs (the
G protein that mediates the stimulation of adenylyl
cyclase) also has been reported to be modulated by
glucocorticoids.
58
As a result,
2
-adrenoceptor-stim
-
ulated adenylyl cyclase activity and cAMP accumu-
lation increase after glucocorticoid treatment.
Effect of Long-Acting
-Agonists on Corticosteroids
In addition to the beneficial effects of corticoste-
roids on LABA activity, LABAs also may enhance the
effects of corticosteroids. It has been observed in
vitro, using primary human lung fibroblasts and
vascular smooth muscle cells, that LABAs induce
glucocorticoid receptor (GR) translocation from the
cell cytosol to the nucleus and enhance GR-glu-
cocorticoid response element binding in the absence
of ligand.
59
Preliminary reports in vivo have con-
firmed this finding.
60
It seems likely, therefore, that
LABAs may be able to induce GR nuclear translo-
cation and that this may prime the receptor to be
more responsive to a concomitant or subsequent
challenge with glucocorticoids (Fig 3). LABAs also
may increase the sensitivity of the molecular path-
ways that are utilized by corticosteroids to suppress
Figure 2. Effects of LABA/ICS combination therapy on pathophysiology of COPD. The LABA/ICS
combination has additive effects on the pathophysiology of COPD affecting airway obstruction,
inflammation, structural changes, and mucociliary dysfunction. It addresses the multicomponent nature
of COPD more than does LABA or ICS treatment alone.
www.chestjournal.org CHEST / 126/1/JULY, 2004 225
inflammation, including their action on histone acet-
ylation and deacetylation,
61,62
or through effects on
the activation of transcription factors, such as nuclear
factor (NF)-B.
63
Recently, it has been docu-
mented
64
that the combination of glucocorticoids
and LABAs synchronizes the activation of the GR
and CCAAT-enhancer binding protein (C/EBP)-,
which belongs to a family of transcription factors that
are involved in the differentiation process of numer-
ous tissues. This action results in a faster and more
prolonged activation of p21(Waf1/Cip1) compared
with each drug given alone. In particular, when
administered alone, formoterol and budesonide ac-
tivate the two transcription factors with different
potency and kinetics. The combined administration
of the two drugs leads to a simultaneous activation of
the GR and C/EBP-, resulting in a synergistic
stimulatory effect on p21(Waf1/Cip1) promoter ac-
tivity. Furthermore, when administered together,
the drugs are effective at concentrations that are
ineffective when either drug is administered alone.
These data lend support to the idea that the com-
bined application of the two drugs has a beneficial
effect that is greater than that of increasing the dose
of either drug alone, at least in asthma.
65,66
Role of LACA/ICS Interactions in COPD
At present, the role of these interactions in COPD
patients cannot be clearly defined. GR does not
interact only with C/EBP-, but also affects the
action of other transcription factors, such as activating
protein 1, cAMP responsive element-binding protein,
and the inhibitor of NF-B.
6770
A direct interaction
with the GR also has been shown for the signal
transducer and activator of transcription 3 and 5.
71,72
These findings suggest that we must also investigate the
possibility of an interaction between glucocorticoids
and LABAs at these levels, and evaluate the potential
impact on COPD management.
Figure 3. The basis for synergy between LABAs and ICSs. Glucocorticoids (GCS) freely diffuse from
the circulation across cell membranes into cells, where they activate the GR. On ligand binding, the
receptor is activated, dissociates from chaperone proteins, and translocates to the nucleus, where it can
bind as a dimer to specific DNA sequences (glucocorticoid response elements [GRE]) upstream of the
start site of transcription. The activated GCS-GR also binds to the
2
-adrenoceptor gene, leading to an
increase in the number of
2
-adrenoceptors in the cell membrane. On the other side, LABA stimulates
2
-adrenoceptors, leading to the priming of the GR and increasing translocation of the receptor into
the nucleus of the cell. The overall response is increased anti-inflammatory activity from a given steroid
dose. The primed GR also may have enhanced actions against other transcription factors such as
NF-B. Besides, LABAs may inhibit NF-B by inducing an increase in IB- levels.
MAPK mitogen-activated protein kinase; PKA protein kinase A; AC adenylate cyclase;
IKK IB kinase coupler; Ub ubiquitin; Pi phosphatidyl inositol.
226 Reviews
In Vitro Interactions of LABA and ICS Useful in
COPD
Examples of interaction between these two classes
of drugs that might be useful in COPD patients
include the synergistic inhibition by glucocorticoids
and LABAs of TNF--induced IL-8 release from
cultured human airway smooth muscle cells
63
and
alveolar macrophages in patients with COPD,
73
and
the capacity of corticosteroids to counteract the
enhancement of LABAs on TNF--induced IL-8
production in cultured human bronchial epithelial
cells.
74
These effects are mediated through the
2
-adrenoceptor into a signal transduction pathway
that does not involve the GR, but other interactions
between glucocorticoid and
2
-agonist cannot be
excluded.
74
The inhibition of transcription factors,
such as NF-B, by -agonists may occur as a result
of an increase in the levels of the inhibitor of NF-B.
One study
75
of monocytic THP-1 cells stimulated
with lipopolysaccharide showed that -agonists
could inhibit TNF- and IL-8 production, and that
the effect was related to increased cytoplasmic con-
centrations of IB-, possibly through its decreased
degradation. Elevated cAMP levels can inhibit NF-
B-mediated gene transcription in human monocyte
and endothelial cells.
76
cAMP regulates cytokine
gene expression by induction of the cAMP-mediated
transcriptional repressor in human thymocytes.
77
The inhibitory effects mediated by induction of the
cAMP-mediated transcriptional repressor may be
related to its ability to bind (ie, mask) a wide range of
cyclic adenosine monophosphate response element
and activating protein-1 motifs and/or its ability
to inactivate certain transcription complexes via
protein-protein interactions.
76
In any case, IL-8 is a
potent chemoattractant and an activator for neutro-
phils, and this may result in a persistent inflamma-
tory cycle by establishing a positive feedback loop.
Reductions in neutrophil number and function could
reduce the severity of disease and the degree of
airflow obstruction in patients with COPD.
63
Another important finding is the capacity of both
ICSs and LABAs to reduce the total number of
bacteria adhering to the respiratory mucosa in a
concentration-dependent manner without altering
the bacterial tropism for mucosa, and to preserve
ciliated cells.
78
ICSs and LABAs, when administered
together at low concentrations, exhibited a synergis-
tic effect with respect to the preservation of ciliated
cells, showing a trend toward reduced damage and a
significant preservation of the number of ciliated
cells compared to either agent alone at the same
concentrations. This result may have clinical signifi-
cance as it is thought that ciliated cells are the most
sensitive to damage by bacterial infection.
79
It is
well-known that airway colonization and chronic
infection contribute to progressive pulmonary dam-
age in COPD patients via the action of proinflam-
matory substances in what is known as the vicious
circle theory
80
(Fig 4). Recently, the synergistic
Figure 4. The LABA/ICS combination breaks the COPD vicious cycle. LABAs can break the COPD
vicious cycle by increasing cilial beat frequency, and therefore enhancing mucus clearance, by
preventing bacterial damage to the epithelium, and by suppressing neutrophil activation and mediator
release. Besides, ICSs and LABAs when administered together at low concentrations exhibited a
synergistic effect with respect to the preservation of ciliated cells, showing a trend toward reduced
damage and a significant preservation of the number of ciliated cells compared to either agent alone
at the same concentrations.
www.chestjournal.org CHEST / 126/1/JULY, 2004 227
effects of salmeterol and fluticasone in human
rhinovirus-induced proinflammatory cytokine pro-
duction have been documented.
81
Rhinoviruses are
implicated in many acute exacerbations of COPD,
perhaps by inducing proinflammatory cytokines.
82
Inhaled Combination Therapy With LABAs
and Corticosteroids in Stable COPD
Patients
Several clinical trials support the concept of in-
haled combination therapy with LABAs and cortico-
steroids in stable COPD patients, although some
reports have not been published yet. Nonetheless,
the currently available information has recently in-
duced the European Agency for the Evaluation of
Medicinal Products to issue a positive opinion
83
on
the use of combination therapy with LABAs and
ICSs in the maintenance treatment of patients with
severe COPD and a history of exacerbations.
Clinical Effects in Adding an ICS to a LABA
The results of a recent small study
84
have indi-
cated that the addition of budesonide amplifies the
fast onset of action of formoterol, but does not
induce systemic effects, in patients with COPD. It
has been shown that glucocorticoids act on steroid
receptors to increase
2
-adrenoceptor expression by
augmenting the rate of
2
-adrenoceptor gene tran
-
scription.
85
However, the glucocorticoid-induced in-
crease in the accumulation of
2
-adrenoceptor mes
-
senger RNA could be detected at 15 min, the
maximal accumulation occurred at 2 h, and the
glucocorticoid-induced increase in
2
-adrenoceptor
messenger RNA returned to the control level by
17 h. In contrast, the increase in the
2
-adrenocep
-
tor values was slower, reaching a maximum between
17 and 24 h. These findings indicate that another
type of interaction may underlie the rapid onset of
action of combination therapy with formoterol and
budesonide. It is likely that a nongenomic action of
budesonide might explain this effect. Thus, it is
possible that corticosteroids might induce a fall in
intracellular Ca
2
that might result, in part, from the
inhibition of Ca
2
entry through voltage-gated ion
channels.
86
The capacity of corticosteroids to prevent homol-
ogous down-regulation of
2
-adrenoceptor values
and to induce an increase in the rate of synthesis of
these receptors may be important when a patient is
receiving regular treatment with LABAs. The addi-
tion of an ICS to a LABA was initially studied in a
3-month trial that enrolled 80 COPD patients. The
association therapy progressively improved lung
function over the 3-month period compared to long-
acting bronchodilator treatment alone, although the
difference was not statistically significant.
87
How-
ever, the association of therapy with salmeterol
(50 g twice daily) with that with fluticasone (250 or
500 g twice daily) allowed a significantly greater
improvement in lung function after salbutamol ther-
apy alone than salmeterol therapy (50 g twice daily)
alone. It is likely that this effect was due to the
prevention of the homologous down-regulation of
2
-adrenoceptors and the induction of an increase in
the rate of synthesis of receptors through a process of
increased
2
-adrenoceptor gene transcription in
-
duced by fluticasone. This may be considered clini-
cally important, because when airway obstruction
becomes more severe the preferred therapeutic op-
tion is to add a fast-acting inhaled
2
-agonist as
rescue medication to produce rapid relief of bron-
chospasm.
Combination Therapy With Salmeterol/Fluticasone
The value of regular combination therapy with
LABAs and corticosteroids delivered via a single
inhaler to COPD patients has been documented
repeatedly (Table 6). Since the overall goals for
improving clinical outcomes are to reduce symp-
toms, especially dyspnea, to improve exercise capac-
ity, to reduce exacerbations and the possible need for
hospitalization, and to enhance health status,
1
the
different trials have not only evaluated the efficacy of
combination therapy on lung function, but also its
impact on these other clinical outcomes.
A 24-week study with the combination therapy of
salmeterol, 50 g twice daily, and fluticasone propi-
onate, 500 g twice daily, explored the potential for
increasing airflow, reducing symptoms (including
dyspnea), and improving health status, compared
with the individual components and placebo.
88
The
results showed that the salmeterol/fluticasone propi-
onate combination not only improved airflow ob-
struction but also provided clinical benefits, as man-
ifested by reduced severity of dyspnea, reduced use
of rescue salbutamol, and improved health status.
A 52-week multicenter, randomized, double-
blind, placebo-controlled trial (Trial of Inhaled Ste-
roids and Long-Acting
2
-Agonists [TRISTAN])
89
compared the safety and efficacy of the salmeterol,
50 g twice daily/fluticasone propionate, 500 g
twice daily, combination with that of the individual
drugs alone in 1,465 patients with COPD (mean
FEV
1
, 45% predicted). Following a year of treat
-
ment with the salmeterol/fluticasone propionate
combination, patients with COPD experienced sig-
nificant and clinically meaningful improvements in
health status, as measured by the St. George Respi-
ratory Questionnaire (SGRQ), compared with pla-
228 Reviews
cebo or fluticasone alone therapy. In addition, pa-
tients treated with combination therapy had greater
reductions in symptom scores compared with all
other treatments, and greater reductions in activity
(limitation) scores compared with placebo and fluti-
casone therapy alone. These findings could have a
real impact on the management of COPD. Patients
with COPD have significant and measurable de-
creases in health status, which worsen with disease
progression. The fatigue and emotional distress re-
sulting from COPD symptoms such as dyspnea,
cough, and phlegm production reduce the ability of
patients to work or carry out their normal activities.
As there is currently no cure for COPD, the goals of
therapy are to prevent symptoms and exacerbations,
to preserve lung function, and to maintain health
status.
The prevention of exacerbations is a key goal for
the effective management of COPD,
1
since exacer-
bations are known to reduce QOL and are cost
drivers in the treatment of COPD patients. There-
fore, controlling exacerbations is important from
both economic and patients perspectives. Subgroup
analyses of the TRISTAN trial
89
have been per-
formed to determine the relative efficacy of the
salmeterol, 50 g twice daily/fluticasone, 500 g
twice daily, combination therapy in reducing exacer-
bations in COPD patients with FEV
1
values of
50% predicted at baseline (ie, the more severe
subgroup) or FEV
1
values of 50% predicted at
baseline (ie, the less severe subgroup). Exacerbations
were defined a priori as a worsening of COPD
symptoms that required treatment with antibiotics,
oral corticosteroids, or both. Episodes that required
corticosteroid treatment or hospital admission were
noted separately. A total of 949 patients (65%) had a
prebronchodilator FEV
1
of 50% predicted and
were classified into the more severe subgroup, and
513 patients (35%) had FEV
1
values of 50%
predicted and were classified into the less severe
Table 6Long-term Effect of Combination Therapy in COPD*
Study
Patients Enrolled, No./Study
Duration
Changes in FEV
1
Over Baseline at
the End of Treatment Health Outcomes
S/FP
Mahler et al
88
691 patients with moderate-
to-severe COPD (FEV
1
,
65% predicted)/F/U at
24 wk
Pretreatment FEV
1
: 156 mL after
S, 50 g/FP, 500 g twice daily,
107 mL after S, 50 g twice
daily; 104 after FP, 500 g
twice daily; 4 mL after placebo
Greater improvements in the transition
dyspnea index with S/FP compared with
FP, S, and placebo; greater increase in
chronic respiratory disease questionnaire
score with S/FP compared with FP and
placebo, but not with S
Calverley et al
89
1,465 patients, with
moderate-to-severe COPD
(mean FEV
1
, 45%
predicted)/F/U at 52 wk
10% after S, 50 g/FP 500, g
twice daily; 2% after S, 50 g
twice daily; 2% after FP, 500
g twice daily; 3% after
placebo
Significantly greater reduced rate in
decline of the disease-specific SGRQ
after S/FP than after FP and placebo;
No. of exacerbations per patient per year
reduced by 25% after S/FP, 20% after S,
and 19% after FP, all vs placebo, but
30% reduction with S/FP vs placebo
when baseline FEV
1
50% predicted,
and 10% reduction when baseline FEV
1
50% predicted
Dal Negro et al
90
18 patients with moderate
COPD (mean FEV
1
,
49.1% predicted)/F/U at
52 wk
6.6% after S, 50 g/FP 250 g
twice daily; 0.3% after S 50 g
twice daily; 2.6% after placebo
Decrease in exacerbations only after S/FP
F/B
Szafranski et al
91
812 patients with moderate-
to-severe COPD (mean
FEV
1
, 36% of predicted)/
F/U at 52 wk
15% after F, 6 g/B, 200 g
twice daily vs placebo; 9% after
F, 6 g/B, 200 g twice daily vs
B, 200 g twice daily; 1% after
F, 6 g/B, 200 g twice daily vs
F, 6 g twice daily
Significantly greater reduced rate in
decline of the disease-specific SGRQ
after F/B than after placebo; No. of
severe exacerbations per patient per year
reduced after F/B by 24% vs placebo,
23% vs F, and 11% vs B
Calverley and
Olsson,
92
Jones and
Ståhl,
93
and
Calverley and
Peterson
94
1,022 patients with
moderate-to-severe COPD
(mean FEV
1
, 36%
predicted)/F/U at 52 wk
14% after F, 12 g/B, 400 g
twice daily vs placebo; 11%
after F, 12 g/B, 400 g twice
daily vs B, 400 g twice daily;
5% after F, 12 g/B, 400 g
twice daily vs F, 12 g twice daily
Significantly greater reduced rate in
decline of the disease-specific SGRQ
after F/B than after F, B, and placebo;
No. of exacerbations per patient per year
reduced by 24% after F/B, 11.6% after
B, and 3% after FP, all vs placebo
*S salmeterol; FP fluticasone propionate; F formoterol; B budesonide. See Table 4 for abbreviation not used in the text.
www.chestjournal.org CHEST / 126/1/JULY, 2004 229
subgroup. During the study, there was a higher
incidence of exacerbations in the more severe sub-
group compared with the less severe subgroup (567
patients [60%] vs 226 patients [44%], respectively).
In the total population, the salmeterol/fluticasone
combination treatment produced a significant reduc-
tion in the exacerbation rate of 25%, compared with
placebo treatment. This reduction was 30% in the
more severe subgroup, compared with a 10% reduc-
tion in the less severe subgroup. These effects of
combination therapy were more pronounced when
considering exacerbations requiring oral corticoste-
roids, which were reduced by 39% in the total
population, 43% in the more severe subgroup, and
24% in the less severe subgroup. Salmeterol/flutica-
sone combination treatment also reduced the num-
ber of exacerbations requiring therapy with oral
corticosteroids by 19%, compared with therapy with
salmeterol alone, in the more severe subgroup.
Although twice-daily therapy with salmeterol/flutica-
sone was effective in significantly reducing the num-
ber of exacerbations across both categories of disease
severity, there was a trend for a larger reduction in
exacerbations and exacerbations requiring oral corti-
costeroids in the more severe subgroup.
Recently, Dal Negro et al
90
have compared ther-
apy with salmeterol, 50 g twice daily/fluticasone,
250 g twice daily, via a single inhaler with therapy
with salmeterol, 50 g twice daily, alone and placebo
in 52-week study that has enrolled a small group of
ICS-naive patients with moderate COPD who had
already been treated with theophylline, 400 mg per
day, and SABAs on demand. The mean ( SD)
number of exacerbations per year decreased from
3.5 0.8 to 1.16 0.75 in the salmeterol/flutica-
sone group (p 0.001), from 3.0 0.89 to
2.3 0.81 in the salmeterol group (difference not
significant), and from 3.16 1.16 to 4.16 0.75 in
the placebo group (difference not significant). Pa-
tients receiving salmeterol/fluticasone therapy
showed the highest mean improvement in FEV
1
(mean increase, 6.6 2.4%) over the baseline pre-
treatment value (p 0.001), with FEV
1
remaining
unchanged after 52 weeks of treatment in the salme-
terol group (mean increase, 0.3 0.9%) and with a
substantial decrease following placebo therapy
(mean decrease, 2.6 0.5; p 0.001).
Combination Therapy With Formoterol/Budesonide
The effects of formoterol/budesonide combination
treatment in COPD patients also have been investi-
gated. In a 12-month, randomized, double-blind,
placebo-controlled, parallel-group, multicenter
study,
91
812 adults with moderate-to-severe COPD
(mean age, 64 years; mean FEV
1
, 36% predicted;
median time since diagnosis, 5 years) received two
inhalations twice daily of formoterol (6 g)/budes-
onide (200 g), budesonide (200 g) alone, formot-
erol (6 g) alone, or placebo. Formoterol/budes-
onide treatment increased FEV
1
by 15% vs placebo,
9% vs budesonide alone, and 1% vs formoterol alone.
Significant improvements in morning and evening
peak expiratory flow (PEF) values were seen during
the 12 months of therapy with formoterol/budes-
onide compared with all other treatment groups
(adjusted mean change from run-in: morning PEF,
24, 16, and 12 L/min vs placebo, budesonide alone,
and formoterol alone, respectively; evening PEF, 20,
15, and 11 L/min vs placebo, budesonide alone, and
formoterol alone, respectively; all p 0.001). These
lung function improvements were maintained
throughout the 12-month study. The improvement
in lung function observed after formoterol/budes-
onide treatment must be considered to be important,
but the greater capacity of formoterol/budesonide to
decrease the mean total symptom score (p 0.001
vs placebo; p 0.001 vs budesonide alone; p 0.103
vs formoterol alone), to increase the number of days
free from shortness of breath by 12% vs placebo
(p 0.001), and to increase the number of awaken-
ing-free nights by 14% vs placebo (p 0.001), each
of which is equivalent to approximately 1 extra
day/night per week, is likely to have a more substan-
tial impact on health status. Formoterol/budesonide
also significantly reduced the use of reliever medi-
cation by 1.3 and 0.7 inhalations per 24 h vs placebo
and budesonide alone, respectively (both p 0.001).
The sustained reduction in overall symptoms, in-
cluding the number of sleep-disturbed nights, in
patients with moderate-to-severe COPD was re-
flected in improvements in health-related QOL.
Mean reductions from baseline were 3.9, 1.9,
3.6 and 0.03, respectively, after therapy with
formoterol/budesonide, budesonide, formoterol, and
placebo.
91
Compared with treatment with placebo,
treatment with formoterol/budesonide significantly
improved the SGRQ total score (p 0.009), and the
symptom domain score (p 0.001) and impact do-
main score (p 0.006). Greater improvements were
seen with formoterol/budesonide compared with the
other active treatments, but these did not achieve
statistical significance.
Formoterol/budesonide therapy also produced
clinically significant reductions in the number of
exacerbations in patients with moderate-to-severe
COPD.
91
It reduced the number of severe exacer-
bations per patient per year by 24% vs placebo
therapy (p 0.035), by 23% vs formoterol therapy
(p 0.043), and by 11% vs budesonide therapy
(p 0.385). Moreover, budesonide/formoterol ther-
apy also reduced the number of mild exacerbations
230 Reviews
compared with therapy with placebo (62%;
p 0.001), budesonide alone (35%; p 0.022); and
formoterol alone (15%; p 0.403).
In another randomized, double-blind, active-con-
trolled study, 1,022 patients (mean age, 64 years;
FEV
1
, 0.99 L and 36% predicted) with moderate-to-
severe COPD and a history of exacerbations had
their condition optimized with therapy with formot-
erol, 12 g twice daily, and oral prednisolone prior to
being given formoterol (12 g twice daily)/budes-
onide (400 g twice daily), budesonide alone (400
g twice daily), formoterol alone (12 g twice daily),
or placebo for 1 year.
9294
The study, which has not
been published yet, was designed to determine
whether these treatments prevented COPD exacer-
bations. Formoterol/budesonide therapy was signifi-
cantly better than that with long-acting
2
-agonists
alone, corticosteroids alone, or placebo in maintain-
ing the lung function improvement achieved after
optimization.
92
Mean FEV
1
in the budesonide/
formoterol group remained 14% above the value in
the placebo group (p 0.001), 11% above the value
in the budesonide group (p 0.001), and 5% above
the value in the formoterol group (p 0.01). Morn-
ing PEF in the budesonide/formoterol group was 18
L/min above the value in the placebo group
(p 0.001), 15 L/min above the value in the budes-
onide group (p 0.001), and 7 L/min above the
value in the formoterol group (p 0.01). The value
for evening PEF in the budesonide/formoterol group
was 14 L/min above the value in the placebo group
(p 0.001), 12 L/min above the value in the budes-
onide group (p 0.001), and 5 L/min above the
value in the formoterol group (p 0.056). For FVC,
the value in the formoterol/budesonide group was
9% above that in the placebo group (p 0.001), 8%
above that in the budesonide group (p 0.001), and
2% above that in the formoterol group. These
changes, which occurred soon after treatment began,
showed no sign of tachyphylaxis over the 12-month
study period. Formoterol/budesonide therapy also
improved SGRQ total score.
93
In particular, it im-
proved all domain scores vs placebo (p 0.01),
activity score by 3.6 vs budesonide therapy
(p 0.05) and 3.5 vs formoterol therapy (p 0.05),
and impact score by 5.7 vs budesonide (p 0.001)
and 3.7 vs formoterol (p 0.05).
Interestingly, formoterol/budesonide therapy pro-
vided significantly better protection against exacer-
bations than either LABA alone, corticosteroid ste-
roid alone, or placebo treatment.
94
It prolonged the
time to the first exacerbation requiring medical
intervention (ie, hospitalization and/or the use of oral
steroids/antibiotics) compared with therapy with
budesonide alone (p 0.05), formoterol alone
(p 0.01), or placebo (p 0.05). The median dura-
tions to the first exacerbation were as follows:
formoterol/budesonide, 254 days; budesonide alone,
178 days; formoterol alone, 154 days; and placebo, 96
days. Formoterol/budesonide therapy also reduced
the risk of a first exacerbation by 29% vs placebo
(p 0.01), 30% vs formoterol alone (p 0.01), and
23% vs budesonide alone (p 0.05). Fewer exacer-
bations per patient per year were recorded in the
formoterol/budesonide group (1.38) than in the
budesonide alone group (1.60), the formoterol-alone
group (1.85), or the placebo group (1.80). The
reduction in the mean exacerbation rate, compared
with placebo, was 11.6% in the budesonide-alone
group (difference not significant), 3.0% in the
formoterol-alone group (difference not significant),
and 24% in the formoterol/budesonide group
(p 0.05). The numerically larger reduction in the
formoterol/budesonide group suggests that synergy
may occur. Furthermore, formoterol/budesonide
treatment significantly reduced the mean number of
oral steroid courses by 45% vs placebo (p 0.001),
28% vs budesonide alone, and 30% vs formoterol
alone (both p 0.05). Budesonide therapy alone
reduced the mean number of oral steroid courses by
23% vs placebo (p 0.05).
Comparison Between Combination Therapy With
Salmeterol/Fluticasone and Formoterol/Budesonide
Apparently, there is no substantial difference be-
tween salmeterol/fluticasone and formoterol/budes-
onide in patients with COPD when these combina-
tions are prescribed at the dosages recommended for
this pathology, at least after acute administration.
95
Systemic Adverse Effects Using
Combination Therapy With LABAs and
Corticosteroids
Consideration of the risk/benefit ratio is important
when using inhaled combination therapy with
LABAs and ICSs in patients with COPD, mainly
because of the potential systemic adverse effects
induced by ICSs.
Side Effects of Long-term Treatments With High-
Dose ICSs
It is well-known that long-term treatments with
high-dose ICSs have the potential to produce sys-
temic adverse effects. Patients with COPD may be
particularly vulnerable to these systemic effects as
they are often elderly, immobile, and have poor
nutrition, thus increasing the risk of osteoporosis.
37
However, the results of different trials have been
variable. In the Lung Health Study,
45
a significantly
www.chestjournal.org CHEST / 126/1/JULY, 2004 231
lower bone density in the lumber spine and femur
was observed, whereas the EUROSCOP study
42
reported no difference between study arms. A recent
Cochrane review
96
of the effects on bone metabo-
lism in COPD patients reported that there is no
evidence of an effect of ICS treatment on bone
mineral density or vertebral fracture when given at
conventional doses for 2 or 3 years. Higher doses
have been associated with biochemical markers of
increased bone turnover, but data on bone mineral
density and the incidence of fractures at these doses
are not available. There is a need for further, even
longer term, prospective studies of conventional and
high doses of ICSs.
Elderly patients also may have an increased risk of
developing cataracts, glaucoma, and diabetes.
37
However, no differences in the rates of cataract were
seen in the most important long-term trials.
4245,49
In
the EUROSCOP study,
42
10% of patients developed
skin bruising compared to 4% in the control group. It
is generally accepted that any discussion of the use of
high-dose ICSs in patients with COPD must weigh
the real risk of systemic side effects against the
minimal clinical value provided by this treatment.
37
Side Effects of Combination Therapy With
Salmeterol/Fluticasone
Although there is clear evidence that the concen-
tration of ICSs can be reduced when combined with
2
-agonists, thus minimizing the risk of side ef
-
fects,
64
there is a real need to establish whether any
side effects induced by combination therapy with
LABAs and ICSs are outweighed by the clinical
advantages.
The combined use of salmeterol and fluticasone
propionate in a single formulation provides additive
benefit in the treatment of COPD but with compa-
rable safety to the individual components used
alone.
88,89
The safety profile of salmeterol/flutica-
sone combination treatment was consistent with that
observed with the administration of salmeterol plus
fluticasone, and was not different from that for the
monocomponents alone after a 12-week treatment
period.
87
The overall adverse event profile of the
combination therapy showed no new or unexpected
adverse events. There was no evidence that combi-
nation treatment with the salmeterol/fluticasone
combination was associated with any increased risk
of clinically relevant hypothalamic-pituitary-adrenal
axis suppression (as assessed by cosyntropin stimula-
tion testing) compared with treatment with flutica-
sone, salmeterol, or placebo alone. No unexpected
cardiovascular effects, as assessed by Holter moni-
toring and routine ECG, were observed in those
patients receiving salmeterol/fluticasone combina-
tion treatment compared with patients receiving
salmeterol or placebo. In the TRISTAN study,
89
all
treatments were well-tolerated, although the per-
centage of patients with oropharyngeal candidiasis
was higher in groups receiving fluticasone-containing
treatment (placebo treatment, 2%; salmeterol treat-
ment alone, 2%; fluticasone treatment alone, 7%;
salmeterol/fluticasone combination treatment, 8%).
The majority of patients (96%) had serum cortisol
levels that were within the reference range or not
significantly changed from baseline. The incidence
of skin bruising was low and was comparable among
treatment groups (placebo group, 6%; salmeterol
alone group, 6%; fluticasone alone group, 7%;
salmeterol/fluticasone combination group, 8%).
ECG findings were unchanged by therapy.
Side Effects of Combination Therapy With
Formoterol/Budesonide
Formoterol/budesonide therapy also was shown to
be well-tolerated and to have a safety profile similar
to that for therapy with placebo and the monocom-
ponents in patients with moderate-to-severe COPD
during 12 months of treatment.
91
Most adverse
events were reported for respiratory system disor-
ders, particularly COPD (formoterol/budesonide
group, 17%; placebo group, 26%; budesonide-alone
group, 13%; and formoterol-alone group, 19%). No
treatment-related patterns were discernible regard-
ing death (26 deaths) or serious adverse events
(number of serious adverse events per 1,000 treat-
ment days: formoterol/budesonide group, 0.8; pla-
cebo group, 0.9; budesonide-alone group, 0.7; for-
moterol-alone group, 0.7). Discontinuations due to
disease deterioration were highest in the placebo
group (placebo group, 21%; formoterol/budesonide
group, 10%; budesonide-alone group, 12%; formot-
erol-alone group, 14%), while the frequency of
discontinuations due to other adverse events was
similar in all groups (6 to 8%). No clinically impor-
tant between-group differences were identified for
laboratory or ECG measurements (including QTc
prolongation).
Effect of LABA/ICS Combination Therapy on
2
-Adrenoceptor Tolerance
These clinical findings regarding combination
treatment with both salmeterol/fluticasone and for-
moterol/budesonide are important. Tolerance gener-
ally develops to systemic
2
-mediated adverse ef
-
fects, but it has been reported
97
that concomitant
therapy with inhaled budesonide resensitized the
response of cardiac
2
-adrenoceptors to salbutamol
in subjects who were receiving regular twice-daily
formoterol therapy, which could suggest an in-
232 Reviews
creased propensity for the development of systemic
2
-mediated adverse effects. However, a previous
study
98
has demonstrated that the addition of the
recommended dose of formoterol to ICS therapy did
not induce significant nonpulmonary consequences,
except tremor, which may sometimes be a limiting
effect.
Need for Further Studies
First of all, there is a need to assess the antiin-
flammatory effects of long-term inhaled combination
therapy with LABAs and ICSs. There is little knowl-
edge at present about the effects of regular treat-
ment with LABAs and/or ICSs on airway inflamma-
tion in COPD patients. Studies
99
using bronchial
biopsy specimens obtained during fiberoptic bron-
choscopy in patients with COPD have yielded valu-
able information about the inflammatory process in
the large airways of patients with this disease. Bron-
choscopic biopsy provides specimens of airway wall
tissue from which tissue morphology can be as-
sessed, inflammatory cells can be quantified, and
gene products can be identified. Consequently, the
immunopathology of bronchial biopsy specimens
could be used to evaluate the antiinflammatory
effects of combination therapy. Obviously, less inva-
sive methods also can be used.
100
Another important point is the need to relate the
short-term response to either ICSs or LABAs to a
long-term benefit with inhaled combination therapy.
This information is extremely important when we
consider that only a positive response to a 2-week
course of oral prednisolone (eg, 30 mg per day) or a
6-week course of ICSs (eg, beclomethasone, 500 g
twice daily, or equivalent) would justify the prescrip-
tion of regular inhaled steroids to COPD patients,
and that a limitation on the use of LABAs to patients
with a demonstrable response to SABAs also has
been recommended.
101
It should be noted that
formoterol/budesonide combination treatment was
effective even in patients who had not been opti-
mized with oral corticosteroid therapy.
93
There is also a clear need for a study to examine
whether adding a LABA to ICS therapy produces
greater improvements in lung function and symptom
control than increasing the ICS dose. On the other
hand, we also need to know whether adding an ICS
to a regular LABA treatment produces greater im-
provements in lung function and symptom control
than increasing the dose of LABA alone. In other
words, we need to know whether more integrated
medical treatment can be more effective and safer
than more aggressive therapy with the individual
agent alone. Obviously, it is also important to inves-
tigate whether such integrated therapy can change
the natural history of COPD.
Information on the economic impact of the various
treatment options in COPD is also required. To date,
few studies have examined the clinical and economic
impact of treatments in COPD patients. One
study
102
has examined the use of a LABA in COPD
patients, and has demonstrated that significant im-
provements in health and QOL may be obtained at a
moderate cost. In another study,
103
early interven-
tion with an ICS resulted in significant health gains
at a relatively low cost. We do not know whether the
higher medication cost of combination therapy with
a LABA and ICS, compared with monotherapy, is
really justified from a pharmacoeconomic point of
view for COPD patients. Looking at the results of
studies that have been published in the literature
or have been presented as abstracts to relevant
international meetings,
8894
it is likely that the higher
cost of medication is almost completely offset by
savings in other costs, such as consultations and
hospital admissions for treating exacerbations. Exac-
erbations are particularly important as they are major
cost drivers, the costs varying considerably with the
severity of the exacerbation. One study
104
found that
2
-agonist therapy in conjunction with corticosteroid
therapy resulted in improved health outcomes at a
small increase in cost compared to
2
-agonist ther
-
apy alone. The addition of an ICS to a
2
-agonist
resulted in net additional health-care costs of $201
per patient-year compared to therapy with a
2
-
agonist alone, but more than half of the additional
cost of adding the ICS was offset by a reduction in
the costs of other health-care services. A recent
health economic analysis
105
comparing formoterol/
budesonide therapy (12/400 g, respectively, bid)
with therapy with formoterol (12 g bid), budes-
onide (400 g bid), and placebo was performed on
data from a 1-year multinational, prospective, ran-
domized, parallel-group clinical study among 1,022
patients with COPD. The difference in total health-
care costs (eg, hospitalizations, emergency depart-
ment visits, general practitioner visits, study drugs,
and concomitant medication use) associated with the
different treatments was analyzed using Swedish unit
costs. Health-care costs were numerically, but not
statistically, significantly lower for formoterol/budes-
onide therapy (22,893 Swedish kronor [SEK] per
year) compared with therapy with formoterol
(33,211 SEK per year), budesonide (28,134 SEK per
year), and placebo (29,207 SEK per year). The
results were robust, as this pattern remained the
same when applying UK unit costs (formoterol/
budesonide therapy, £1,595 per year; formoterol,
£2,577 per year; budesonide, £2,748 per year; pla-
cebo, £2,044 per year), and when analyzing sub-
www.chestjournal.org CHEST / 126/1/JULY, 2004 233
groups of severe patients (FEV
1
40% predicted)
and European patients separately.
Conclusions
There is an increasing volume of evidence showing
that combination treatment with both salmeterol/
fluticasone and formoterol/budesonide can provide
an effective treatment option for COPD patients.
However, it must always be considered that the trials
that explored the effect of these therapies recruited
a select group of patients with COPD, and the
results may not be generalizable to patients with
extremely advanced or mild forms of the disorder.
106
Nevertheless, since the goals of COPD therapy are
to control symptoms, to prevent exacerbations, and
to improve lung function and health status, the
efficacy of combination therapy in inducing long-
lasting bronchodilation, improving QOL, and pre-
venting exacerbations must be considered a real
advancement in a disease for which treatment re-
mains suboptimal for a considerable number of
patients.
The capacity of combination treatment to reduce
exacerbations is particularly important. As correctly
stressed by Calverley and coauthors,
89
the low rate of
acute episodes might be attributable to regression to
the mean in the number of exacerbations or an effect
of improved care associated with clinical trials, but
suggests that a study of longer duration and with a
larger number of participants would be needed to
show a difference. Such long-term studies are un-
derway. They will allow us to better evaluate the
effect of such combination therapy on disease pro-
gression and mortality, and to determine the long-
term tolerability of combination therapy in patients
with COPD.
References
1 Pauwels RA, Buist AS, Calverley PMA, et al. Global strategy
for the diagnosis, management, and prevention of chronic
obstructive pulmonary disease: NHLBI/WHO Global Initia-
tive for Chronic Obstructive Lung Disease (GOLD) work-
shop summary. Am J Respir Crit Care Med 2001; 163:1256
1276
2 Saetta M, Turato G, Maestrelli P, et al. Cellular and
structural bases of chronic obstructive pulmonary disease.
Am J Respir Crit Care Med 2001; 163:1304 1309
3 Fabbri LM, Romagnoli M, Corbetta L, et al. Differences in
airway inflammation in patients with fixed airflow obstruc-
tion due to asthma or chronic obstructive pulmonary dis-
ease. Am J Respir Crit Care Med 2003; 167:418 424
4 Barnes PJ. Chronic obstructive pulmonary disease. N Engl
J Med 2000; 343:269 280
5 Cazzola M, Spina D, Matera MG. The use of bronchodila-
tors in stable chronic obstructive pulmonary disease. Pulm
Pharmacol Ther 1997; 10:129 144
6 Cazzola M, Donner CF. Long-acting
2
agonists in the
management of stable chronic obstructive pulmonary dis-
ease. Drugs 2000; 60:307320
7 Dahl R, Greefhorst LAPM, Nowak D, et al. Inhaled formot-
erol dry powder versus ipratropium bromide in chronic
obstructive pulmonary disease. Am J Respir Crit Care Med
2001; 164:778 784
8 Wadbo M, Lofdahl CG, Larsson K, et al. Effects of formot-
erol and ipratropium bromide in COPD: a 3-month placebo-
controlled study. Eur Respir J 2002; 20:11381146
9 Mahler DA, Donohue JF, Barbee RA, et al. Efficacy of
salmeterol xinafoate in the treatment of COPD. Chest 1999;
115:957965
10 Rennard SI, Anderson W, ZuWallack R, et al. Use of a
long-acting inhaled
2
-adrenergic agonist, salmeterol xin
-
afoate, in patients with chronic obstructive pulmonary dis-
ease. Am J Respir Crit Care Med 2001; 163:10871092
11 Siafakas NM, Vermeire P, Pride NB, et al. Optimal assess-
ment and management of chronic obstructive pulmonary
disease (COPD): a consensus statement of the European
Respiratory Society. Eur Respir J 1995; 8:13981420
12 van Noord JA, de Munck DR, Bantje TA, et al. Long-term
treatment of chronic obstructive pulmonary disease with
salmeterol and the additive effect of ipratropium. Eur
Respir J 2000; 15:878 885
13 ZuWallack RL, Mahler DA, Reilly D, et al. Salmeterol plus
theophylline combination therapy in the treatment of
COPD. Chest 2001; 119:16611670
14 DUrzo AD, De Salvo MC, Ramirez-Rivera A, et al. In
patients with COPD, treatment with a combination of
formoterol and ipratropium is more effective than a combi-
nation of salbutamol and ipratropium: a 3-week, random-
ized, double-blind, within-patient, multicenter study. Chest
2001; 119:13471356
15 Johnson M, Rennard S. Alternative mechanisms for long-
acting
2
-adrenergic agonists in COPD. Chest 2001; 120:
258270
16 Bloeman PGM, Van den Tweek MC, Henricks PAJ, et al.
Increased cAMP levels in stimulated neutrophils inhibit
their adhesion to human bronchial epithelial cells. Am J
Physiol 1997; 272:L580L587
17 Eda R, Tanimoto Y, Marou H, et al. Comparison of effect of
formoterol and salbutamol on human neutrophil function in
vitro. Allergy 1997; 91:301
18 Ottonello L, Morone P, Dapino P, et al. Inhibitory effect of
salmeterol on the respiratory burst of adherent human
neutrophils. Clin Exp Immunol 1996; 106:97102
19 Ward C, Li X, Wang N, et al. Salmeterol reduces BAL IL-8
levels in asthmatics on low dose inhaled corticosteroids
[abstract]. Eur Respir J 1998; 12:380S
20 Dentener MA, Buurman WA, Wouters EFM. Theophylline
and the -agonists salmeterol and salbutamol potently block
neutrophil activation, as indicated by a reduced release of
bacterial/permeability-increasing protein [abstract]. Am J
Respir Crit Care Med 1998; 157:A602
21 Johnson M. Effects of
2
-agonists on resident and infiltrat
-
ing inflammatory cells. J Allergy Clin Immunol 2002; 110:
S282S290
22 Bowden JJ, Sulakvelidze I, McDonald DM. Inhibition of
neutrophil and eosinophil adhesion to venules of rat trachea
by
2
-adrenergic agonist formoterol. J Appl Physiol 1994;
77:397405
23 Davies A, Lefkowitz R. Agonist-promoted high affinity state
of -adrenergic receptor in human neutrophils: modulation
by corticosteroids. J Clin Endocrinol Metab 1981; 53:
703708
234 Reviews
24 Saari SM, Vidgren MT, Herrala J, et al. Possibilities of
formoterol to enhance the peripheral lung deposition of the
inhaled liposome corticosteroids. Respir Med 2002; 96:999
1005
25 Thompson AB, Mueller MB, Heires AJ, et al. Aerosolized
beclomethasone in chronic bronchitis: improved pulmonary
function and diminished airway inflammation. Am Rev
Respir Dis 1992; 146:389 395
26 Keatings VM, Jatakanon A, Worsdell YM, et al. Effects of
inhaled and oral glucocorticoids on inflammatory indices in
asthma and COPD. Am J Respir Crit Care Med 1997;
155:542548
27 Culpitt SV, Maziak W, Loukidis S, et al. Effect of high dose
inhaled steroid on cells, cytokines, and proteases in induced
sputum in chronic obstructive pulmonary disease. Am J
Respir Crit Care Med 1999; 160:16351639
28 Loppow D, Schleiss MB, Kanniess F, et al. In patients with
chronic bronchitis a four week trial with inhaled steroids
does not attenuate airway inflammation. Respir Med 2001;
95:115121
29 Lomas DA, Ip M, Chamba A, et al. The effect of in vitro and
in vivo dexamethasone on human neutrophil function.
Agents Actions 1991; 33:279 285
30 Llewellyn-Jones CG, Hill SL, Stockley RA. Effect of fluti-
casone propionate on neutrophil chemotaxis, superoxide
generation, and extracellular proteolytic activity in vitro.
Thorax 1994; 49:207212
31 Llewellyn-Jones CG, Harris TA, Stockley RA. Effect of
fluticasone propionate on sputum of patients with chronic
bronchitis and emphysema. Am J Respir Crit Care Med
1996; 153:616 621
32 Confalonieri M, Mainardi E, Della Porta R, et al. Inhaled
corticosteroids reduce neutrophilic bronchial inflammation
in patients with chronic obstructive pulmonary disease.
Thorax 1998; 53:583585
33 Yildiz F, Kaur AC, Ilgazli A, et al. Inhaled corticosteroids
may reduce neutrophilic inflammation in patients with
stable chronic obstructive pulmonary disease. Respiration
2000; 67:7176
34 Gizycki MJ, Hattotuwa KL, Barnes N, et al. Effects of
fluticasone propionate on inflammatory cells in COPD: an
ultrastructural examination of endobronchial biopsy tissue.
Thorax 2002; 57:799 803
35 Hattotuwa KL, Gizycki MJ, Ansari TW, et al. The effects of
inhaled fluticasone on airway inflammation in chronic
obstructive pulmonary disease: a double-blind, placebo-
controlled biopsy study. Am J Respir Crit Care Med 2002;
165:15921596
36 Balbi B, Majori M, Bertacco S, et al. Inhaled corticosteroids
in stable COPD patients: do they have effects on cells and
molecular mediators of airway inflammation? Chest 2000;
117:16331637
37 Barnes PJ. Inhaled corticosteroids are not beneficial in
chronic obstructive pulmonary disease. Am J Respir Crit
Care Med 2000; 161:342344
38 Calverley PM. Inhaled corticosteroids are beneficial in
chronic obstructive pulmonary disease. Am J Respir Crit
Care Med 2000; 161:341342
39 Chanez P, Vignola AM, OShaugnessy T, et al. Corticoste-
roid reversibility in COPD is related to features of asthma.
Am J Respir Crit Care Med 1997; 155:1529 1534
40 Pizzichini E, Pizzichini MM, Gibson P, et al. Sputum
eosinophilia predicts benefit from prednisone in smokers
with chronic obstructive bronchitis. Am J Respir Crit Care
Med 1998; 158:15111517
41 Nishimura K, Koyama H, Ikeda A, et al. The effect of
high-dose inhaled beclomethasone dipropionate in patients
with stable COPD. Chest 1999; 115:3137
42 Pauwels RA, Lo¨fdahl CG, Laitinen LA, et al. Long-term
treatment with inhaled budesonide in persons with mild
chronic obstructive pulmonary disease who continue smok-
ing: European Respiratory Society Study on Chronic Ob-
structive Pulmonary Disease. N Engl J Med 1999; 340:
19481953
43 Vestbo J, Sorensen T, Lange P, et al. Long-term effect of
inhaled budesonide in mild and moderate chronic obstruc-
tive pulmonary disease: a randomized controlled trial. Lan-
cet 1999; 353:18191823
44 Burge PS, Calverley PM, Jones PW, et al. Randomised,
double blind, placebo controlled study of fluticasone propi-
onate in patients with moderate to severe chronic obstruc-
tive pulmonary disease: the ISOLDE trial. BMJ 2000;
320:12971303
45 Lung Health Study Research Group. Effect of inhaled
triamcinolone on the decline in pulmonary function in
chronic obstructive pulmonary disease. N Engl J Med 2000;
343:19021909
46 Suissa S. Effectiveness of inhaled corticosteroids in chronic
obstructive pulmonary disease: immortal time bias in obser-
vational studies. Am J Respir Crit Care Med 2003; 168:
4953
47 Pauwels R. Inhaled glucocorticosteroids and chronic ob-
structive pulmonary disease: how full is the glass? Am J
Respir Crit Care Med 2002; 165:1579 1580
48 Thompson WH, Carvalho P, Souza JP, et al. Controlled trial
of inhaled fluticasone propionate in moderate to severe
COPD. Lung 2002; 180:191201
49 Paggiaro PL, Dahle R, Bakran I, et al. Multicentre random-
ised placebo-controlled trial of inhaled fluticasone propi-
onate in patients with chronic obstructive pulmonary dis-
ease. Lancet 1998; 351:773780
50 Alsaeedi A, Sin DD, McAlister FA. The effects of inhaled
corticosteroids in chronic obstructive pulmonary disease: a
systematic review of randomized placebo-controlled trials.
Am J Med 2002; 113:59 65
51 Sin DD, Tu JV. Inhaled corticosteroids and the risk of
mortality and readmission in elderly patients with chronic
obstructive pulmonary disease. Am J Respir Crit Care Med
2001; 164:580 584
52 OBrien A, Russo-Magno P, Karki A, et al. Effects of
withdrawal of inhaled steroids in men with severe irrevers-
ible airflow obstruction. Am J Respir Crit Care Med 2001;
164:365371
53 van der Valk P, Monninkhof E, van der Palen J, et al. Effect
of discontinuation of inhaled corticosteroids in patients with
chronic obstructive pulmonary disease: the COPE Study.
Am J Respir Crit Care Med 2002; 166:1358 1363
54 OBrien A, Ward NS. Steroid therapy in chronic obstructive
pulmonary disease. Med Health R I 2002; 85:5255
55 Barnes PJ. Efficacy of inhaled corticosteroids in asthma.
J Allergy Clin Immunol 1998; 102:531538
56 Lipworth BJ. Airway subsensitivity with LABAs: is there
cause for concern? Drug Saf 1997; 16:295308
57 Schramm CM. -Adrenergic relaxation of rabbit tracheal
smooth muscle: a receptor deficit that improves with corti-
costeroid administration. J Pharmacol Exp Ther 2000; 292:
280287
58 Mak JCW, Nishikawa M, Shirasaki H, et al. Protective
effects of a glucocorticoid on downregulation of pulmonary
2
-adrenergic receptors in vivo. J Clin Invest 1995; 96:
99106
www.chestjournal.org CHEST / 126/1/JULY, 2004 235
59 Eickelberg O, Roth M, Lorx R, et al. Ligand-independent
activation of the glucocorticoid receptor by
2
-adrenergic
receptor agonists in primary human lung fibroblasts and
vascular smooth muscle cells. J Biol Chem 1999; 274:1005
1010
60 Adcock IM, Maneechotesuwan K, Usmani O. Molecular
interactions between glucocorticoids and long-acting
2
-
agonists. J Allergy Clin Immunol 2002; 110(suppl):S261
S268
61 Ito K, Barnes PJ, Adcock IM. Glucocorticoid receptor
recruitment of histone deacetylase 2 inhibits IL-1-induced
histone H4 acetylation on lysines 8 and 12. Mol Cell Biol
2000; 20:6891 6903
62 Ito K, Jazwari E, Cosio B, et al. p65-activated histone
acetyltransferase activity is repressed by glucocorticoids:
mifepristone fails to recruit HDAC2 to the p65/HAT com-
plex. J Biol Chem 2001; 276:30208 30215
63 Pang L, Knox AJ. Synergistic inhibition by
2
-agonists
and corticosteroids on tumor necrosis factor induced
interleukin-8 release from cultured human airway smooth
muscle cells. Am J Respir Cell Mol Biol 2000; 23:79 85
64 Roth M, Johnson PR, Rudiger JJ, et al. Interaction between
glucocorticoids and
2
agonists on bronchial airway smooth
muscle cells through synchronised cellular signalling. Lancet
2002; 360:12931299
65 Woolcock A, Lundback B, Ringdahl N, et al. Comparison of
addition of salmeterol to inhaled steroids with doubling of
the dose of inhaled steroids. Am J Respir Crit Care Med
1996; 153:14811488
66 Pauwels RA, Lofdahl CG, Postma DS, et al. Effect of
inhaled formoterol and budesonide on exacerbations of
asthma. N Engl J Med 1997; 337:14051411
67 Jonat C, Rahmsdorf HJ, Park KK, et al. Antitumor promo-
tion and antiinflammation: down-modulation of AP-1 (Fos/
Jun) activity by glucocorticoid hormone. Cell 1990; 62:
11891204
68 Heck S, Bender K, Kullmann M, et al. IB-independent
down regulation of NF-B activity by glucocorticoid recep-
tor. EMBO J 1997; 16:4698 4707
69 Sheppard KA, Phelps KM, Williams AJ, et al. Nuclear
integration of glucocorticoid receptor and nuclear factor-B
signaling by CREB-binding protein and steroid receptor
coactivator-1. J Biol Chem 1998; 273:2929129294
70 Strahle U, Schmid W, Schutz G. Synergistic action of the
glucocorticoid receptor with transcription factors. EMBO J
1988; 7:3389 3395
71 Zhang Z, Jones S, Hagood JS, et al. STAT3 as a co-activator
of glucocorticoid receptor signaling. J Biol Chem 1997;
272:3060730610
72 Stocklin E, Wissler M, Gouilleux F, et al. Functional
interactions between Stat5 and the glucocorticoid receptor.
Nature 1996; 383:726728
73 Seeto LJ, Burgess JK, Johnson PR, et al. Effect of flutica-
sone and salmeterol on human alveolar macrophage cyto-
kine production in patients with chronic obstructive pulmo-
nary disease (COPD) [abstract]. Am J Respir Crit Care Med
2003; 167:A318
74 Korn SH, Jerre A, Brattsand R. Effects of formoterol and
budesonide on GM-CSF and IL-8 secretion by triggered
human bronchial epithelial cells. Eur Respir J 2001; 17:
10701077
75 Farmer P, Pugin J. -adrenergic agonists exert their anti-
inflammatory effects in monocytic cells through the IkB/
NF-kB pathway. Am J Physiol 2000; 279:L675L682
76 Ollivier V, Parry GCN, Cobb RR, et al. Elevated cyclic AMP
inhibits NF-kB-mediated transcription in human monocytic
cells and endothelial cells. J Biol Chem 1996; 271:20828
20835
77 Bodor J, Habener JF. Role of transcriptional repressor
ICER in cyclic AMP-mediated attenuation of cytokine gene
expression in human thymocytes. J Biol Chem 1998; 273:
95449551
78 Dowling RB, Johnson M, Cole PJ, et al. Effect of fluticasone
propionate and salmeterol on Pseudomonas aeruginosa in-
fection of the respiratory mucosa in vitro. Eur Respir J 1999;
14:363369
79 Tsang KW, Rutman A, Tanaka E, et al. Interaction of
Pseudomonas aeruginosa with human respiratory mucosa
in vitro. Eur Respir J 1994; 7:17461753
80 Wilson R. Infections of the airways. Curr Opin Infect Dis
1991; 4:166 177
81 Edwards MR, Johnson M, Johnston SL. Synergistic effects
of salmeterol and fluticasone in human rhinovirus induced
pro-inflammatory cytokine production [abstract]. Am J
Respir Crit Care Med 2003; 167:A399
82 Seemungal T, Harper-Owen R, Bhowmik A, et al. Respira-
tory viruses, symptoms, and inflammatory markers in acute
exacerbations and stable chronic obstructive pulmonary
disease. Am J Respir Crit Care Med 2001; 164:16181623
83 European Committee for Proprietary Medicinal Products.
Press release: European Agency for the Evaluation of Medic-
inal Products; Committee for Proprietary Medicinal Products
meeting of 21 to 23 January 2003. Available at: http://www.
emea.eu.int/pdfs/human/press/pr/029403en.pdf. Accessed June
14, 2004
84 Cazzola M, Santus P, Di Marco F, et al. Onset of action of
budesonide/formoterol in single inhaler vs. formoterol in
patients with COPD [abstract]. Am J Respir Crit Care Med
2003; 167:A318
85 Mak JC, Nishikawa M, Barnes PJ. Glucocorticosteroids
increase
2
-adrenergic receptor transcription in human
lung. Am J Physiol 1995; 12;L41L46
86 Borski RJ. Nongenomic membrane actions of glucocorti-
coids in vertebrates. Trends Endocrinol Metab 2000; 11:
427436
87 Cazzola M, Di Lorenzo G, Di Perna F, et al. Additive effects
of salmeterol and fluticasone or theophylline in COPD.
Chest 2000; 118:15761581
88 Mahler DA, Wire P, Horstman D, et al. Effectiveness of
fluticasone propionate and salmeterol combination deliv-
ered via the Diskus device in the treatment of chronic
obstructive pulmonary disease. Am J Respir Crit Care Med
2002; 166:1084 1091
89 Calverley P, Pauwels R, Vestbo J, et al. Combined salmet-
erol and fluticasone in the treatment of chronic obstructive
pulmonary disease: a randomised controlled trial. Lancet
2003; 361:449 456
90 Dal Negro RW, Pomari C, Tognella S, et al. Salmeterol &
fluticasone 50 g/250 g bid in combination provides a
better long-term control than salmeterol 50 g bid alone
and placebo in COPD patients already treated with theoph-
ylline. Pulm Pharmacol Ther 2003; 16:241246
91 Szafranski W, Cukier A, Ramirez A, et al. Efficacy and safety
of budesonide/formoterol in the management of chronic
obstructive pulmonary disease. Eur Respir J 2003; 21:7481
92 Calverley PMA, Olsson H. Budesonide/formoterol in a
single inhaler sustains improvements in lung function over
12 months compared with monocomponents and placebo in
patients with COPD [abstract]. Am J Respir Crit Care Med
2003; 167:A319
93 Jones PW, Ståhl E. Budesonide/formoterol in a single
inhaler improves health status in patients with COPD
[abstract]. Am J Respir Crit Care Med 2003; 167:A320
236 Reviews
94 Calverley PMA, Peterson S. Combining budesonide/
formoterol in a single inhaler reduces exacerbation fre-
quency in COPD [abstract]. Am J Respir Crit Care Med
2003; 167:A948
95 Cazzola M, Santus P, di Marco F, et al. Combination therapy
with salmeterol fluticasone and formoterol budesonide
in patients with COPD. Respir Med 2003; 97:453 457
96 Jones A, Fay JK, Burr M, et al. Inhaled corticosteroid effects
on bone metabolism in asthma and mild chronic obstructive
pulmonary disease. Cochrane Database Syst Rev 2002;
1:CD003537
97 Aziz I, McFarlane LC, Lipworth BJ. Concomitant inhaled
corticosteroid resensitises cardiac
2
-adrenoceptors in the
presence of long-acting
2
-agonist therapy. Eur J Clin
Pharmacol 1998; 54:377381
98 Centanni S, Carlucci P, Santus P, et al. Non-pulmonary
effects induced by the addition of formoterol to budesonide
therapy in patients with mild or moderate persistent asthma.
Respiration 2000; 67:60 64
99 Hattotuwa K, Gamble EA, OShaughnessy T, et al. Safety of
bronchoscopy, biopsy, and BAL in research patients with
COPD. Chest 2002; 122:1909 1912
100 Rytila PH, Laitinen A, Lindqvist A, et al. Comparison
between bronchial biopsy and induced sputum in patients
with chronic bronchitis and chronic obstructive pulmonary
disease [abstract]. Am J Respir Crit Care Med 1999;
159:A515
101 COPD Guidelines Group of the Standards of Care Com-
mittee of the BTS. BTS guidelines for the management of
chronic obstructive pulmonary disease. Thorax 1997;
52(suppl):S1S28
102 van den Boom G, Rutten-van Molken MP, Molema J, et al.
The cost effectiveness of early treatment with fluticasone
propionate 250 g twice a day in subjects with obstructive
airway disease: results of the DIMCA program. Am J Respir
Crit Care Med 2001; 164:20572066
103 Jones PW, Wilson K, Sondhi S. Cost-effectiveness of sal-
meterol in patients with chronic obstructive pulmonary
disease: an economic evaluation. Respir Med 2003; 97:
2026
104 Rutten-Van Molken MP, Van Doorslaer EK, Jansen MC,
et al. Cost and effects of inhaled corticosteroids and bron-
chodilators in asthma and chronic obstructive pulmonary
disease. Am J Respir Crit Care Med 1995; 151:975982
105 Lo¨ fdahl C-G, Andreasson E, Svensson K, et al. Budesonide/
formoterol in a single inhaler improves overall health status
in patients with COPD without increasing healthcare costs
[abstract]. Am J Respir Crit Care Med 2003; 167:A89
106 Hackam DG. Treatment of chronic obstructive pulmonary
disease: combination or component therapy? Can Med
Assoc J 2003; 168:1296 1297
www.chestjournal.org CHEST / 126/1/JULY, 2004 237
... A solid pharmacological rationale supports the combination of an ICS with a LABA in COPD [34]. LABAs produce both bronchodilator and non-bronchodilator effects, whereas ICSs are potent anti-inflammatory agents [34]. ...
... A solid pharmacological rationale supports the combination of an ICS with a LABA in COPD [34]. LABAs produce both bronchodilator and non-bronchodilator effects, whereas ICSs are potent anti-inflammatory agents [34]. This means that ICSs and LABAs treat different aspects of COPD and their combination has the potential to induce a greater benefit in the management of COPD. ...
Article
Introduction Triple therapy in COPD is becoming increasingly important with the cumulative documentation of its ability to reduce the risk of AECOPD. However, it must be established which patients benefit most from it compared to other treatments. Areas covered We critically review the literature to determine, if possible, the real role of triple therapy in the treatment of COPD. We have identified studies from several databases and selected the information thought to be more significant. Expert opinion It is still unclear whether and when addition of an ICS to the LAMA/LABA combination provides real additional clinical value, regardless of a preventive effect on exacerbations. There are many doubts about the value of the blood eosinophil count as a valid biomarker to predict AECOPD risk and the clinical response to ICS, also because no association was found in observational studies. In any case, before starting a therapy involving ICS, the risk factors for the development of pneumonia must always be evaluated. Adding a LAMA to an ICS/LABA combination seems to be less problematic. However, each LABA/LAMA combination has a specific efficacy/safety profile that needs to be considered for personalized therapy in COPD even in the context of triple therapy.
... COPD patients who scored positive in the reversibility test should only be treated with ICS without the need for additional LABA. It has also been noted that the combination of ICS and LABA in a same single inhaler has enhanced lung functions and decreased the severity of dyspnea in COPD patients [40] . The aforementioned conclusions are all supported and published by the TORCH study [39] . ...
Article
Full-text available
Short acting beta2-agonists (SABAs) such as salbutamol and terbutaline are commonly used in the treatment of asthma and chronic obstructive pulmonary disease (COPD). According to the guideline produced by the British Thoracic Society (BTS) and the Scottish Intercollegiate Guidelines Network (SIGN), SABAs are considered to be the first line of treatment in asthma management. The National Institute for Health and Clinical Excellence (NICE) guidelines also recommend the use of SABA as the first line of treatment for managing patients with COPD. The recommendations in NICE guidelines state that in COPD patients, breathlessness and exercise limitation should be first medicated with short-acting bronchodilators (including beta2-agonists or anticholinergics) when it is required. Both salbutamol and terbutaline have been widely used for asthma and COPD, and they have been found to be extremely beneficial in the two pathologic conditions.
... Several mechanisms have been suggested to explain the synergy between ICSs, LAMAs and LABAs. Bidirectional molecular interactions between corticosteroids and β 2 -agonists cause the pharmacological effects of ICSs and LABAs to be enhanced reciprocally [6,7]. Corticosteroids increase the expression of β 2 -adrenoceptors (β 2 -ARs) on the cell surface by increasing the transcription of the gene of these receptors. ...
Article
Full-text available
Introduction: : Single inhaler triple therapy (SITT) with an inhaled corticosteroid, a long-acting β2-agonist, and a long-acting muscarinic antagonist is an effective and attractive therapeutic option codified in the recommendations of guidelines and treatment strategies for the management of COPD. Areas covered: : The preclinical and clinical development in COPD of fluticasone furoate (FF)/umeclidinium (UMEC)/vilanterol (VI) SITT and its use in the real world. Expert opinion: : Findings from phase III/IV trials and the use of FF/UMEC/VI in the real-world setting support the view that it may be a useful, safe, and cost-effective option for the maintenance treatment of COPD, especially when dealing with patients who are not adequately controlled with dual ICS/LABA or LAMA/LABA therapy. Only direct head-to-head comparisons will be able to establish whether FF/UMEC/VI may be preferable to the other SITTs approved for COPD due to its pharmacokinetic and pharmacodynamic characteristics and especially the fact that it is the only one that can be taken once-daily. In addition, there is a need for further studies, especially in the real world, to optimize the positioning of FF/UMEC/VI in the treatment of COPD, also considering the availability of FF/VI and UMEC/VI and the need for better differentiation between the three treatments.
... Важное место среди ЛС, использующихся для дли тельной поддерживающей терапии ХОБЛ, занимают комбинированные препараты -иГКС плюс длитель но действующие β 2 агонисты (ДДБА) [17][18][19]. Также как иГКС, комбинированная терапия иГКС / ДДБА в настоящее время рекомендована для применения пациентам с III-IV стадиями ХОБЛ (по классифика ции GOLD) и с частыми обострениями [1]. Научное обоснование комбинации иГКС и ДДБА вытекает из комплиментарных эффектов препаратов: иГКС по вышают экспрессию гена β 2 рецептора и уменьшают потенциал для развития десенситизации β 2 рецепто ра, в то время как β 2 агонисты активируют неактив ные ГКС рецепторы, делая их более чувствитель ными для стероидозависимой активации [20][21][22]. ...
... The working mechanisms of bronchodilators are to reduce airflow obstruction and decrease respiratory symptoms by reducing dynamic hyperinflation. [71] I.3.2. Treatment for post-9/11 lung injury ...
Thesis
Treatment response is the result of a complex interaction between host factors, environmental factors and drug-related factors. Those factors were studied separately in order to obtain the best possible picture of the optimal response to inhaled corticosteroids (ICS). First, we characterized the type of lung injury of World Trade Center (WTC)-exposed firefighters, identifying individuals and their characteristics. Host factors associated with prolonged ICS-containing treatment were body mass index, lower post-9 September 2001 (9/11) spirometry and first post-9/11 modified Medical Research Council (mMRC) dyspnea score. Worse baseline forced expiratory volume in 1 second (FEV1) and respiratory symptoms are host factors that predict response to ICS. Regarding biomarkers, a higher blood eosinophil count and increase in IL- 4 were associated with an increased risk of irritant-induced airflow obstruction. Furthermore, low immunoglobulin A (IgA) soon after WTC exposure was associated with lower longitudinal FEV1 measurements, increased risk of airflow obstruction and increased antibiotic treatment. Moreover, low IgA tends to predict response to ICS. Second, the environmental factor associated with prolonged ICS/LABA treatment was high intensity WTC exposure defined by arrival morning of 9/11. Last, drug-related factors associated with the response to ICS treatment were assessed. In a longitudinal study, ICS treatment improved lung function decline in individuals with WTC-related lung injury. This improvement was more pronounced when WTC-exposed firefighters were more adherent to ICS. Pharmaceutical counseling interventions, analyzed through nation-wide pharmacy data, led to a marked improvement in the adherence of asthma patients to ICS. This also resulted in an improved controller-to-total-ratio, indicating an improvement in asthma control. In conclusion, improved adherence to ICS treatment leads to reduced lung function decline and better asthma control.
... For several years, there has been evidence of a pharmacological advantage in combining LABAs and ICSs in both US RESPIRATORY AND PULMONARY DISEASES asthma 3 and COPD. 4 In effect, bidirectional molecular interactions between corticosteroids and β 2 -agonists are responsible for a mutual enhancement of their pharmacological effects. 5 The interaction between these drugs at the level of human isolated bronchi has previously been investigated; results showed that LABAs and ICSs synergistically relax airways to an extent that is very strong at low-to-medium concentrations mainly in passively sensitized small airways, a validated ex vivo model of bronchial asthma. ...
... ICS overuse might have several explanations: i) evidence coming from specific randomized controlled trials that are generalized to the whole population of COPD [69,70,85,86,88]; ii) the concept that COPD is still considered an inflammatory disease in which the airway inflammation can be effectively modulated by an anti-inflammatory drug; iii) the addictive/ synergistic pharmacological effect to long-acting bronchodilators, especially LABAs [89,90]; iv) a sort of 'copy and paste' to COPD of the positive ICS effects observed in asthma [91]; and v) the inappropriate differential diagnosis between the two obstructive pulmonary diseases [77]. ...
Article
Full-text available
Introduction: In 2011, the GOLD recommendations for the treatment of Chronic Obstructive Pulmonary Disease (COPD) introduced new clinical elements to classify the severity of the disease and to guide pharmacological choice. For the first time in the GOLD documents, treatment decision was no longer guided only by pulmonary function, but by a more complex combination of pulmonary function and clinical aspects. The recent versions of the GOLD recommendations introduce new aspects for the clinicians and pose new question for the management of the disease. In addition, inflammatory biomarkers and blood eosinophil levels, have been considered to guide treatment selection. Area covered: The evolution of disease management proposed by the GOLD document opens several areas of debate. A series of roundtable discussions among respiratory physicians took place in Italy to address key clinical questions. Particularly, the role of lung function and the use of biomarkers, the adherence to international guidelines and the possibility to personalize the pharmacological approach in COPD patients have been discussed, summarized and analyzed. Expert opinion: The authors believe that the development of a precision medicine approach tailoring the specific treatment for each patient is the goal of COPD management and may be achieved by considering the phenotypic classification of COPD patients.
Article
The claimed functional basis for ICSs in asthma and COPD is airway selectivity, attained by inhaling a potent, lipophilic compound with long local dissolution/absorption time. The development has been empirically based, resulting in five widely used ICSs. Among them, budesonide (BUD) deviates by being less lipophilic, leading to a more rapid systemic uptake with plasma peaks with some systemic anti-inflammatory activity. By this, BUD fits less well into the current pharmacological dogma of optimal ICS profile. In this review we compared the physicochemical, pharmacological and clinical properties of BUD, fluticasone propionate (FP) and fluticasone furoate (FF), representing different levels of lipophilicity, airway and systemic kinetics, focusing on their long-acting β2-agonist (LABA) combinations, in line with current GINA and GOLD recommendations. We are aware of the differences between formoterol (FORM) and the not rapid acting LABAs such as e.g. salmeterol and vilanterol but our comparisons are based on currently available combination products. A beclomethasone dipropionate (BDP)/FORM combination is also commented upon. Based on clinical comparisons in asthma and COPD, we conclude that the BUD/formoterol (BUD/FORM) combination is as effective and safe as the FP and FF combinations, and is in some cases even better as it can be used as “maintenance plus reliever therapy” (MART) in asthma and as maintenance in COPD. This is difficult to explain by current views of required ICS's/LABAs pharmacokinetic profiles. We propose that BUD achieves its efficacy by a combination of airway and systemic activity. The airway activity is dominating. The systemic activity contributes by plasma peaks, which are high enough for supportive anti-inflammatory actions at the blood and bone marrow levels but not sufficiently long to trigger a similar level of systemic adverse effects. This may be due to BUD's capacity to exploit a systemic differentiation mechanism as programmed for cortisol's various actions. This differentiation prospect can be reached only for an ICS with short plasma half-life. Here we present an alternative mode for an ICS to reach combined efficacy and safety, based on a poorly investigated and exploited physiological mechanism. A preference of this mode is broader versatility, due to that its straighter dose-response should allow a better adaptation to disease fluctuations, and that its rapid activity enables use as “anti-inflammatory reliever”.
Chapter
Bronchodilators are the mainstay of the current management of COPD and are critical in the symptomatic management of asthma. Three classes of bronchodilators (β2-AR agonists, mAChR antagonists, and xanthines) are currently available. The combination of two or more drugs with dissimilar but complementary mechanisms of action, of which at least one is a bronchodilator, has become standard in the treatment of asthma and COPD. However, they often do not relieve symptoms and their long-term use raises safety concerns. Therefore, there is a considerable amount of research being undertaken to improve the existing classes of bronchodilators with the development of novel β2-AR agonists, mAChR antagonists, and xanthines. Research is also ongoing to develop new options for combination therapy such as, bifunctional bronchodilator drugs and bifunctional bronchodilator/anti-inflammatory drugs. However, the limits that characterize the existing classes of bronchodilators have maintained a lively interest in identifying new therapeutic options to induce bronchodilation that could help to overcome these limits. Although novel classes have proved difficult to develop, in recent years new targets have been identified. These new opportunities are mostly in a preclinical phase.
Article
The objective of this study was to determine the costs and effects of combined bronchodilator and anti-inflammatory therapy. In a 2.5-yr randomized controlled study, combined beta 2-agonist/corticosteroid therapy (BA + CS) and combined beta 2-agonist/anticholinergic therapy (BA + AC) were compared with beta 2-agonist/placebo therapy (BA + PL). Included in the study were 274 patients 18 to 60 yr of age with moderately severe obstructive airways disease. The main clinical endpoints were lung function, hyperresponsiveness, restricted activity days, and symptom-free days. The economic endpoints were the costs of health care utilization. Compared with BA + PL, BA + CS led to significant improvements in FEV1, PC20, and symptom-free days. BA + AC did not differ from BA + PL in this respect. The respective annual acquisition costs of BA + CS, BA + AC, and BA + PL were 532 US$, 277 US$, and 156 US$. Thus, BA + CS costs 376 US$ more than BA + PL. However, compared with BA + PL therapy, BA + CS led to statistically significant savings in other health care costs of about 175 US$ (95% CI from 46 to 303 US$). Thus, more than half of the additional costs of adding the inhaled corticosteroid are compensated for by a reduction in the costs of other health care services. Overall, inhaled corticosteroids lead to a small but net increase in health care costs of 201 US$ per patient per year.(ABSTRACT TRUNCATED AT 250 WORDS)
Article
Background: Chronic obstructive pulmonary disease (COPD) results from a progressive decline in lung function, which is thought to be the consequence of airway inflammation. We hypothesized that antiinflammatory therapy with inhaled corticosteroids would slow this decline. Methods: We enrolled 1116 persons with COPD whose forced expiratory volume in one second (FEV1) was 30 to 90 percent of the predicted value in a 10-center, placebo-controlled, randomized trial of inhaled triamcinolone acetonide administered at a dose of 600 microg twice daily. The primary outcome measure was the rate of decline in FEV1 after the administration of a bronchodilator. The secondary outcome measures included respiratory symptoms, use of health care services, and airway reactivity. In a substudy of 412 participants, we measured bone density in the lumbar spine and femur at base line and one and three years after the beginning of treatment. Results: The mean duration of follow-up was 40 months. The rate of decline in the FEV1 after bronchodilator use was similar in the 559 participants in the triamcinolone group and the 557 participants in the placebo group (44.2+/-2.9 vs. 47.0+/-3.0 ml per year, P= 0.50). Members of the triamcinolone group had fewer respiratory symptoms during the course of the study (21.1 per 100 person-years vs. 28.2 per 100 person-years, P=0.005) and had fewer visits to a physician because of a respiratory illness (1.2 per 100 person-years vs. 2.1 per 100 person-years, P=0.03). Those taking triamcinolone also had lower airway reactivity in response to methacholine challenge at 9 months and 33 months (P=0.02 for both comparisons). After three years, the bone density of the lumbar spine and the femur was significantly lower in the triamcinolone group (P < or = 0.007). Conclusions: Inhaled triamcinolone does not slow the rate of decline in lung function in people with COPD, but it improves airway reactivity and respiratory symptoms and decreases the use of health care services for respiratory problems. These benefits should be weighed against the potential long-term adverse effects of triamcinolone on bone mineral density.
Article
COPD is a prevalent disease, with an increasing attributable mortality. Because inflammation plays a significant role in the pathogenesis of this disease, the use of anti-inflammatory therapies would appear indicated; hence the widespread use of corticosteroids in COPD. Although the majority of patients with stable COPD do not benefit from systemic steroids, there is good evidence supporting the use of short courses of systemic steroids for COPD exacerbations. With respect to inhaled corticosteroids, the studies are conflicting. Those patients with an asthmatic component to their disease, or with a positive bronchodilator test, appear to benefit most from inhaled steroids. Those with irreversible disease do not benefit from short-term inhaled steroids. Long-term inhaled corticosteroids, though not having a significant effect on the rate of decline in spirometric indices, do appear to decrease the number of exacerbations and the rate of decline in health status, reduce respiratory symptoms, decrease use of health care services, and improve airway reactivity. These effects appear more marked in patients with moderate-to-severe disease. Because very few therapies offer significant benefits to patients with COPD, and until a test is developed that will distinguish between potential steroid responders from non-responders, it is worthwhile giving all patients with COPD a trial (3-6 months) of inhaled corticosteroids to determine whether they are responsive.
Article
Background: Although both inhaled and oral corticosteroids have anti-inflammatory effects causing improvement in clinical symptoms and spirometry in the treatment of asthma, the role of corticosteroids in the management of chronic obstructive pulmonary disease (COPD) is controversial. Objective: To evaluate the effects of inhaled corticosteroids on sputum neutrophilia in clinically stable COPD patients. Methods: In total, 18 patients were enrolled in the study. During 2 months, 9 patients in group A inhaled fluticasone propionate (FP) 500 μg 3 times daily. In group B 9 patients received placebo. All of the patients continued to inhale both salbutamol and ipratropium bromide. In 9 patients, sustained-released theophylline was also administered. Blood samples, spirometric tests, blood gas analyses, and either spontaneous or induced sputum cultures were evaluated on entry into the study, after a 2 months of treatment and following the 6-week washout period. Results: After the 2-month FP treatment, no significant changes in the number of peripheral blood neutrophils, blood gas and spirometry data were observed in both groups. In group A, the total cell number and the number of neutrophils decreased from a mean of 3.4 ± 1.3 × 106 cells/g and 0.6 ± 0.3 × 106 neutrophils/g on entry into study to 1.9 ± 0.6 × 106 cells/g and 0.02 ± 0.01 × 106 neutrophils/g after 8-week treatment with FP, returning to 3.3 ± 1.1 × 106 cells/g and 0.5 ± 0.3 × 106 neutrophils/g following the washout period. The percentages of neutrophils were 55.6 and 77.9% in groups A and B after 2 months of FP treatment. There was no significant change in group B values during the study. Conclusion: These data suggest that neutrophilic inflammation in sputum may be decreased by inhaled corticosteroids in clinically stable COPD patients.
Article
beta (2)-Adrenergic agonists are commonly used as bronchodilators to treat patients with COPD. In addition to prolonged bronchodilation, long-acting beta (2)-agonists (LABAs) exert other effects that may be of clinical relevance. These include inhibition of airway smooth-muscle cell proliferation and inflammatory mediator release, as well as nonsmooth-muscle effects, such as stimulation of mucociliary transport, cytoprotection of the respiratory mucosa, and attenuation of neutrophil recruitment and activation. This review details the possible alternative mechanisms of action of the LABAs, salmeterol and formoterol, in COPD.