Sie sind auf Seite 1von 14

STAT E O F T H E A RT R E V I E W

Asthma-COPD overlap syndrome:


pathogenesis, clinical features, and
therapeutic targets
Janice M Leung, Don D Sin
Division of Respiratory Medicine
and Centre for Heart Lung A B S T RAC T
Innovation, St Paul’s Hospital,
University of British Columbia,
Asthma-COPD overlap syndrome (ACOS) or asthma-COPD overlap captures the
Vancouver, BC, Canada subset of patients with airways disease who have features of both asthma and
Correspondence to: D Sin
don.sin@hli.ubc.ca chronic obstructive pulmonary disease (COPD). Although definitions of ACOS vary, it
Cite this as: BMJ 2017;358:j3772 is generally thought to encompass persistent airflow limitation in a patient older than
doi: 10.1136/bmj.j3772
40 years of age with either a history of asthma or large bronchodilator reversibility.
Series explanation: State of the
Art Reviews are commissioned ACOS affects about a quarter of patients with COPD and almost a third of patients
on the basis of their relevance to
academics and specialists in the US
who previously had asthma. Compared with their counterparts with asthma or COPD
and internationally. For this reason alone, patients with ACOS have significantly worse respiratory symptoms, poorer
they are written predominantly by
US authors quality of life, and increased risk of exacerbations and hospital admissions. Whether
this condition emerges after gradual shifts in airway remodelling and inflammation
in a patient with COPD, as the result of noxious exposures in a patient with asthma,
or even as a de novo disease with its own pathology is yet to be determined.
Nevertheless, using treatments developed for asthma or COPD that target
eosinophilic, neutrophilic, or paucigranulocytic airway inflammation may be a helpful
approach to these patients until further clinical trials can be performed.

Introduction and neutrophilic mediated inflammation, proteolysis,


In 1961 at a bronchitis symposium in Groeningen, the genetics, and childhood lung development and how these
Netherlands, Orie and Sluiter proposed what has since factors might lead to ACOS. Although much remains to be
become known as the Dutch hypothesis, a paradigm determined about any of these factors in the development
under which asthma and chronic obstructive pulmo‑ of ACOS, they provide a framework of understanding as
nary disease (COPD) share a common origin with their to how features of asthma might develop in patients
divergence explained by an individual’s unique genetic with COPD and vice versa. The latter part of the review
make-up and environmental exposures.1 Despite the will then use this framework to speculate on treatments
lasting influence of this paradigm, it was and continues that may benefit patients with ACOS and ultimately to
to be vigorously contested,2‑4 and, in reality, clinicians establish priorities for the development of novel ACOS
have practised for decades under the rubric of a fixed therapeutics.
dichotomy between asthma and COPD. Only recently
has a more fluid understanding of these two conditions Rationale
re-emerged, existing instead along a continuum with The concept of ACOS is controversial, and even the use
a subset of patients showing features of both. Asthma- of the term “syndrome” may meet with some resistance
COPD overlap syndrome (ACOS) or asthma-COPD overlap as we do not yet know whether this is a single, unifying
is now thought to affect up to a quarter of patients previ‑ disease.9 We urge the medical community, however, to
ously thought to have COPD and up to a third of patients consider that many patients do not fall neatly within the
previously thought to have asthma.5‑8 categories of asthma and COPD and that many have a
In this review, we present the case for ACOS as a clini‑ mix of features from both. Importantly, multiple studies
cally important phenotype among airways diseases and have now shown that, compared with those with asthma
advocate for the continued pursuit of its underlying or COPD alone, these patients face increased symptom
pathophysiology. Our main focus is the potential mecha‑ burdens and higher rates of hospital admission and exac‑
nisms of disease, whether ACOS is merely an evolution erbation,10‑16 indicating that ACOS is a distinct phenotype
from longstanding COPD or asthma or is its own disease in the airways disease spectrum and may have its own
entity with a unique pathogenesis. In particular, we eval‑ clinical trajectory. Unfortunately, treatments specifically
uate the effects of smoking, air pollution, eosinophilic targeted at this mixed phenotype are noticeably lack‑

For personal use only 1 of 14


STAT E O F T H E A RT R E V I E W

ing. Patients with ACOS are often overlooked or simply exposures and the development of COPD. We propose that
excluded in clinical trials investigating novel treatments cigarette smoking or exposure to biomass fuel should be
for asthma and COPD.17 18 In addition, the paucity of data an integral component of the definition of ACOS, to avoid
on the mechanistic drivers of ACOS and how these might including patients with asthma who have developed air‑
differ from those of asthma or COPD has limited the dis‑ way remodelling and fixed airflow limitation. Given the
covery of ACOS specific treatments. As a result, how to lack of an accepted gold standard for the diagnosis of
identify and manage these patients remains a puzzle for ACOS, none of the diagnostic schemes in fig 1 has been
clinicians. Investigation of this subset of patients with officially validated.
airways disease is imperative. Of lesser importance in the diagnostic algorithm are
biomarkers commonly used in the diagnosis and manage‑
Sources and selection criteria ment of asthma. Although helpful in identifying COPD
We obtained references through a PubMed search inclu‑ patients with possible asthma overlap, immunoglobulin
sive of publications from 2000 to March 2017. Search E (IgE), sputum or peripheral blood eosinophil count,
terms included “asthma COPD overlap syndrome”, and fractional exhaled nitric oxide (FeNO) are not suf‑
“ACOS”, and “mixed asthma COPD phenotype”. We ficiently sensitive or specific on their own for diagnosis
added additional references on the basis of the reference of ACOS. Elevated IgE concentrations are found in up to
lists of recent review papers, including the 2016 consen‑ half of patients with COPD,24 25 and eosinophilic sub‑
sus statement from Sin et al.17 We retrieved randomised types of COPD are well described in the literature.26‑30
controlled trials, observational cohort studies, systematic The debate about which level of peripheral eosinophil
reviews, meta-analyses, and case series and reports for count to ascribe to ACOS, whether above 2%, 3%, 5%,
further review. We prioritised larger randomised con‑ or instead using an absolute measurement, has also
trolled trials, observational cohort studies, and meta- not been resolved.21‑33 A FeNO cut-off of above 35 ppb
analyses that were published most recently. We filtered may correlate with a post-bronchodilator FEV1 response
search results for relevance to definition, epidemiology, greater than 200 mL and atopy in patients with COPD,25 34
mechanisms, and treatment. We considered only Eng‑ but the few studies that have looked at FeNO in ACOS
lish language publications. For the section on emerging seem to offer conflicting evidence of the test’s discrimina‑
treatments, we searched Clinicaltrials.gov for studies on tory power.6 35 Moreover, like sputum eosinophil measure‑
ACOS, neutrophilic asthma, and eosinophilic COPD. ments,36 37 FeNO is neither widely available at all sites
nor standardised across instruments or centres.38 39 These
Definition and diagnosis factors remain adjunctive tools in the diagnosis of ACOS
Given the nascence of ACOS, no single accepted defini‑ until further studies can be performed.
tion for ACOS exists. Although it coined the term ACOS in
its 2015 update on asthma management and prevention, Epidemiology and clinical features
the Global Initiative for Asthma (GINA) and the Global Without a standard definition or diagnostic system in
Initiative for Chronic Obstructive Lung Disease (GOLD) place, the fact that population estimates of ACOS are
declined to put forth an overarching definition, citing highly variable comes as no surprise. Epidemiological
the need for better phenotyping and further mechanistic studies have used definitions as disparate as the major-
work.19 More recently, the American Thoracic Society and minor criteria system, self reported physician diagnoses,
the National Heart, Lung, and Blood Institute issued a and combinations of spirometry results and symptom
workshop statement concluding that ACOS should not descriptions. Estimates of ACOS in the general popula‑
be considered a discrete disease entity, but rather an tion, using either self reported physician diagnoses or
airways disease phenotype of mixed features.9 Nonethe‑ a combination of airflow obstruction on spirometry and
less, GINA/GOLD identified diagnostic clues that suggest symptom report, fall roughly between 2% and 3%.12‑46 In
ACOS, including persistent yet reversible airflow limita‑ comparison, estimates of asthma and COPD in these same
tion (post-bronchodilator forced expiratory volume in populations tend to be higher, around 5-17% for asthma
1 s (FEV1) to forced vital capacity (FVC) ratio of <70% and 2-12% for COPD. However, the prevalence of ACOS
and FEV1 improvement of >12% and >400 mL from base‑ may vary according to geographic region, with estimates
line after bronchodilator therapy); a history of asthma ranging anywhere from 0.61% in China to 3.7% in the
diagnosed by a doctor, atopy, allergies, or exposure to United States.7 47 In cohorts of COPD patients specifically,
noxious agents; either sputum neutrophilia or eosino‑ the prevalence of ACOS ranges from 6% to 55%,6‑58 with
philia; and age 40 years or older. Other groups, working pooled estimates from a meta-analysis suggesting a preva‑
largely through consensus opinion, have offered systems lence of just over 25%.5 In cohorts of asthma patients, the
of major and minor criteria by which to identify patients prevalence ranges from 10% to 31%.7‑59 In studies looking
who may have ACOS (fig 1).17‑23 Although subtleties exist at patients with any airway obstruction (defined by either
between these proposed systems, they share several key patient report or diagnosis in the medical record of asthma
obligatory features: that patients should be 40 years or or COPD or through spirometry), the prevalence ranges
older, have persistent airflow obstruction, and a history of from 15% to 56%.11‑62 Moving forward, comparisons of
asthma or evidence of bronchodilator reversibility. Curi‑ prevalence across regions and time will ultimately require
ously, many of these systems have not featured environ‑ a standardised definition for ACOS. Nevertheless, no mat‑
mental exposures (either cigarette smoke or biomass fuel) ter which definition is used, ACOS seems to account for a
as a key component despite the clear links between such considerable portion of all airways diseases.

For personal use only 2 of 14


STAT E O F T H E A RT R E V I E W

&,7$7,21 0$-25&5,7(5,$ 0,125&5,7(5,$ ',$*126,62)$&26

6ROHU&DWDOX³D
 &23'SOXV &23'SOXV PDMRUFULWHULD

9HU\SRVLWLYHEURQFKRGLODWRUWHVWşLHLQFUHDVHLQ 7RWDOKLJK,J( 25
)(9ƝDQGƝP/ 3HUVRQDOKLVWRU\RIDWRS\
6SXWXPHRVLQRSKLOLD 3RVLWLYHEURQFKRGLODWRUWHVWşLHLQFUHDVHLQ 
PDMRUDQG
+LVWRU\RIDVWKPD )(9ƝDQGƝP/RYHUEDVHOLQHRQƝ PLQRUFULWHULD

RFFDVLRQV

.REOL]HN
 &23'SOXV &23'SOXV 
PDMRUFULWHULD
6WURQJO\SRVLWLYHEURQFKRGLODWRUWHVWşLHLQFUHDVH 0LOGO\SRVLWLYHEURQFKRGLODWRUWHVWşLH 25
LQ)(9!DQG!P/ LQFUHDVHLQ)(9!DQG!P/
0HWKDFKROLQHFKDOOHQJHWHVWSRVLWLYLW\ (OHYDWHG,J( 
PDMRUDQG
)H12ƝSSEDQGRUVSXWXPHRVLQRSKLOV! +LVWRU\RIDWRS\ 
PLQRUFULWHULD
+LVWRU\RIDVWKPD

&RVLR
 &23'SOXV &23'SOXV PDMRUFULWHULD

+LVWRU\RIDVWKPD ,J(!,8 25
%URQFKRGLODWRUUHVSRQVHWRVDOEXWDPRO!DQG +LVWRU\RIDWRS\
P/ 7ZRVHSDUDWHGEURQFKRGLODWRUUHVSRQVHVWR 
PLQRUFULWHULD
VDOEXWDPRO!DQGP/
%ORRGHRVLQRSKLOV!

6LQ
 &23'SOXV &23'SOXV 
PDMRUFULWHULD
3HUVLVWHQWDLUƠRZOLPLWDWLRQ SRVWEURQFKRGLODWRU 'RFXPHQWHGKLVWRU\RIDWRS\RUDOOHUJLFUKLQLWLV $1'
)(9)9&RU//1LQSDWLHQWVƝ\HDUVRIDJH %URQFKRGLODWRUUHYHUVLELOLW\RI)(9ƝP/
ƝSDFN\HDUVRIWREDFFRVPRNLQJ25HTXLYDOHQW DQGIURPEDVHOLQHRQƝYLVLWV 
PLQRUFULWHULD
LQGRRURURXWGRRUDLUSROOXWLRQH[SRVXUH 3HULSKHUDOEORRGHRVLQRSKLOFRXQWRIƝ
'RFXPHQWHGKLVWRU\RIDVWKPDEHIRUH\HDUVRI FHOOVP/
DJH25EURQFKRGLODWRUUHYHUVLELOLW\!P/LQ)(9

&DWDOGR
 $&26LQD&23'SDWLHQW $&26LQD&23'SDWLHQW PDMRUFULWHULD

+LJKGHJUHHRIYDULDELOLW\LQDLUZD\REVWUXFWLRQ 3HUVRQDORUIDPLO\KLVWRU\RIDWRS\DQGRU,J( $1'
RYHUWLPH)(9YDULDWLRQƝP/ VHQVLWLYLW\WRRQHRUPRUHDLUERUQHDOOHUJHQV
+LJKGHJUHHRIUHVSRQVHWREURQFKRGLODWRUV! (OHYDWHGEORRGRUVSXWXPHRVLQRSKLOVRU PLQRUFULWHULRQ

P/DQGDERYHEDVHOLQH LQFUHDVHG)H12
$VWKPDGLDJQRVHGEHIRUHWKHDJHRI
6\PSWRPYDULDELOLW\
$JH LQIDYRXURIDVWKPD

$&26LQDQDVWKPDSDWLHQW $&26LQDQDVWKPDSDWLHQW
3HUVLVWHQFHRYHUWLPHRIDLUƠRZREVWUXFWLRQ /DFNRIUHVSRQVHRQDFXWHEURQFKRGLODWRUWHVW
)(9)9&RU//1 5HGXFHGOXQJGLƷXVLRQFDSDFLW\
([SRVXUHWRQR[LRXVSDUWLFOHVRUJDVHVZLWKƝ /LWWOHYDULDELOLW\LQDLUZD\REVWUXFWLRQ
SDFN\HDUVIRUVPRNHUV $JHLQIDYRXURI&23' !\HDUV
3UHVHQFHRIHPSK\VHPDRQFKHVW&7VFDQ

Fig 1 |  Consensus diagnostic criteria for asthma-COPD overlap syndrome (ACOS) in patients with airflow limitation. COPD=chronic obstructive pulmonary disease;
CT=computed tomography; FeNO=fractional exhaled nitric oxide; FEV1=forced expiratory volume in 1 s; FVC=forced vital capacity; LLN=lower limit of normal

Many case-control studies comparing ACOS with tro-oesophageal reflux disease, osteoarthritis, osteopo‑
asthma and COPD suggest a specific demographic rosis, depression, and anxiety.42‑63 Importantly, despite
type: patients with ACOS tend to be younger,5‑64 to be divergent definitions and methods, these studies con‑
female,56 63 and to have higher body mass index,5‑64 sistently show a distinct clinical trajectory for ACOS,
lower socioeconomic status,41 42 and lower education one whose severity seems to exceed that of asthma or
levels.41 42 ACOS patients also carry a higher burden of COPD alone. By various disease control measures—
comorbidities, particularly for conditions such as gas‑ pulmonary function,15‑66 respiratory symptoms,43‑67

For personal use only 3 of 14


STAT E O F T H E A RT R E V I E W

Table 1 | Lung function decline in asthma-COPD overlap syndrome (ACOS)


Mean
duration of
Citation No Definition of ACOS follow-up FEV1 decline*
Fu, 201362 Asthma 8 Respiratory symptoms; increased 4 years Asthma 140 (230)
COPD 36 airflow variability; post- COPD 100 (130)
bronchodilator FEV1/FVC<70% and
ACOS 55 ACOS 60 (190)
FEV1<80%
de Marco, Neither 5659 Diagnosis of asthma or asthma-like 9 years Neither 26.2 (31.1-21.3)
201540 Asthma 941 symptoms + pre-bronchodilator Asthma 25.3 (30.5, 20.1)
chronic airflow obstruction,
COPD 166 COPD 37.3 (44, 30.6)
symptoms, history of active
ACOS 218 smoking or occupational exposure ACOS 25.9 (32.2, 19.6)
to vapours, dust, gas, or fumes
Lange, Asthma 124 Self reported asthma + post- 18 years Asthma 26 (29)
201668 COPD 303 bronchodilatory FEV1/FVC<0.70 COPD 46 (28)
ACO with early onset asthma† 62 ACO with early onset asthma 31 (37)
ACO with late onset asthma‡ 188 ACO with late onset asthma 51 (38)
Tkacova, COPD 4453 COPD + positive methacholine 5 years Difference in FEV1 decline between ACOS and
201648 ACOS 1434 challenge test COPD=−10.3 mL/year
COPD=chronic obstructive pulmonary disease; FEV1=forced expiratory volume in 1 s; FVC=forced vital capacity.
*Measured as mL/year with standard deviation or interquartile range, except for Fu et al, which showed results as change in FEV1 over entire four year period.
†Defined as onset before age 40 years.
‡Defined as onset after age 40 years.

exacerbation rates,14‑68 use of respiratory drugs,43‑69 patients with late onset asthma (onset over the age of
overall health status,7‑43 quality of life,5‑55 and disabil‑ 40 years) had the greatest FEV1 decline (49.6 mL/year
ity12‑61—ACOS patients have greater decrements than versus 27.3 mL/year in ACOS patients with early onset
their counterparts with asthma or COPD alone. Specifi‑ asthma and 39.5 mL/year in COPD patients).68 However,
cally, as high as a 20% decrement in per cent predicted other studies have found that FEV1 decline in ACOS was
FEV1 has been shown in ACOS patients compared with similar to that in asthma but better than in COPD,40 or
asthma patients,41 43 and up to a 10% difference has been that no difference existed between ACOS, asthma, and
shown between ACOS and COPD patients.43 Exacerbation COPD groups.62 Similarly, studies have shown increased
rates are up to four to five times higher in ACOS patients mortality,45‑68 decreased mortality,14 73 and no difference
compared with asthma and COPD patients.43 58 Emer‑ in mortality55 56 when comparing ACOS with asthma and
gency department visits,41‑61 hospital admissions,12‑61 COPD. Discrepancies in case definition and variable fol‑
and healthcare use7‑61 are also significantly higher in low-up times may account for these discordant results;
ACOS patients, who incur twice the health related costs in any case, the natural history of ACOS is still very much
of asthma and COPD patients,70 71 mainly through out‑ a question to be answered.
patient visits and drugs.72 The rate of physician visits,
for example, is approximately 1.3 to 1.5 times higher in Mechanisms of disease
ACOS patients than in COPD patients.7 8 The clinical presentation of ACOS in a manner that
Data on excess decline in lung function (table 1) exceeds the expected disease burdens of either asthma
and excess mortality (table 2) in ACOS remain mixed. or COPD alone raises an important question as to its ori‑
In a cohort of asthma, COPD, and ACOS patients, ACOS gin: is ACOS the manifestation of a unique pathogenic

Table 2 | Survival and mortality in asthma-COPD overlap syndrome (ACOS)


Duration of
Citation No Definition of ACOS follow-up Survival and mortality statistics
Diaz-Guzman, Asthma 709 Self report of both asthma and COPD 18 years Odds ratio (95% CI) for death: asthma 3.70 (2.67 to 5.13);
201145 COPD 815 COPD 3.50 (2.52 to 4.85); ACOS 7.64 (5.08 to 11.49)
ACOS 357
Yamauchi, Asthma 19 865 Admission for asthma exacerbation + history of COPD OR admission for COPD Hospital In-hospital mortality: asthma 1.2%; COPD 9.7%; ACOS 2.3%
201573 COPD 4261 exacerbation + history of asthma stay
ACOS 6279
Sorino, 201655 Asthma 159 Post-bronchodilator FEV1/FVC<LLN + history of asthma or very positive 15 years 15 year survival: asthma 47%; COPD 25%; ACOS 35%
COPD 205 bronchodilator test (increase in FEV1≥15% and ≥400 mL) + personal history of
atopy + personal history of wheezing
ACOS 118
Wurst, 201656 COPD 1483 Self report of asthma in COPD patients Not Mortality rate: COPD 10%; ACOS 9%
ACOS 493 available
Tkacova, COPD 4453 COPD + positive methacholine challenge test 5 years Respiratory related mortality rate: COPD 0.77%; ACOS 1.69%
201648 ACOS 1434
Bai, 201714 COPD 65 Post-bronchodilator FEV1/FVC<70% + either smoking ≥10 pack years or long term Median: 45 Mortality rate: COPD 20%; ACOS 4.6%
ACOS 65 exposure to inhaled dust + positive bronchodilator test (improvement of months
FEV1≥12% and 200 mL)
COPD=chronic obstructive pulmonary disease; FEV1=forced expiratory volume in 1 s; FVC=forced vital capacity; LLN=lower limit of normal.

For personal use only 4 of 14


STAT E O F T H E A RT R E V I E W

+2:0,*+7$67+0$%(&20($&26"

$VWKPDWLFDLUZD\ $VWKPDZLWK&23'IHDWXUHV
(RVLQRSKLOV B,/,/,/$ 1HXWURSKLOV
0DVWFHOOV B7/57/5 &'7FHOOV
,J( &LJDUHWWHVPRNH '1$PHWK\ODWLRQ *REOHWFHOOPHWDSODVLD
*REOHWFHOOPHWDSODVLD $LUSROOXWLRQ 2[LGDWLYHVWUHVV 6PRRWKPXVFOH
6PRRWKPXVFOH B003DQG K\SHUWURSK\
K\SHUWURSK\ SURWHRO\VLV 7KLQƠDPPDWLRQ
7KLQƠDPPDWLRQ (PSK\VHPD

,6$&26,762:1',6($6("

*HQHWLF
VXVFHSWLELOLW\

$&26
(DUO\FKLOGKRRG
OXQJGHYHORSPHQW
)(9

$JH

Fig 2 |  Possible mechanisms for the development of asthma-COPD overlap syndrome (ACOS). Two mechanisms by which ACOS might develop are presented.
According to the Dutch hypothesis, asthma and chronic obstructive pulmonary disease (COPD) exist along a continuum with a shared origin. ACOS may fit along
this continuum if, for instance, a patient with asthma is exposed to the usual inhalants that cause COPD, such as cigarette smoke or air pollution. In both humans
with asthma and mouse models of asthma, these exposures lead to neutrophilic inflammation, cytokine release, oxidative stress, DNA methylation changes,
and matrix metalloproteinase mediated proteolysis. Such changes may then shift the asthmatic airway to features of COPD, including fixed airways obstruction
and emphysema. Conversely, the British hypothesis considers asthma and COPD to be unique diseases. Accordingly, ACOS may have its own origins in genetic
predisposition or early childhood events. Whereas single nucleotide polymorphisms in CSMD1 and GPR65 show marginal genome-wide significance for ACOS
compared with COPD alone, recent data suggest that attenuated lung development in early childhood may contribute to future development of ACOS

mechanism, is it simply the additive (or synergistic) result its place alongside asthma and COPD in the single disease
of two distinct pathologies combined together, or is it the continuum, emerging only through the confluence of spe‑
end result of various environmental stimuli and insults cific patient related and environmental factors. Under this
applied to a patient already affected by asthma or COPD? scheme, one could consider these three diseases as being
To answer this question, revisiting the principles of the somewhat in flux, that asthma leading to ACOS and COPD
Dutch hypothesis (and, conversely, its longstanding rival leading to ACOS are two potential pathways by which
the British hypothesis) may be helpful. Fundamentally, ACOS can develop. The British hypothesis as applied to
the Dutch hypothesis posits that asthma and COPD are a ACOS would instead suggest that this is in fact its own
single disease of diffuse airway obstruction (termed by disease with a unique genetic and molecular profile
Orie and Sluiter as “chronic non-specific lung disease”), distinguishing it from asthma and COPD. Suffice to say,
but that both endogenous and exogenous factors help to neither of these theories has yet been definitively proven
steer patients into distinguishable phenotypes that we or disproven when considering the conundrum of ACOS.
understand better today as asthma or COPD.74 These fac‑ We provide evidence that might support either side, but
tors may include, but are certainly not limited to, genet‑ ultimately the message here is that further mechanistic
ics, sex, age, allergens, smoking, air pollution, biomass work is desperately needed to unravel the pathogenesis
fuel, concomitant pulmonary diseases, and infections. On of ACOS.
the other hand, the British hypothesis maintains separate
origins for asthma and COPD, underscored by the fact How might asthma become ACOS?
that each disease has its own characteristic inflammatory The gradual shift of a patient with asthma to one with
profile, genetic polymorphisms, response to treatment, features of both asthma and COPD requires several
and clinical course.2 4 physiological adjustments to occur. Whereas asthma is
In this section, we consider ACOS within the prism of conventionally viewed as a disease of variable airflow
these two schools of thought (fig 2). If the Dutch hypoth‑ obstruction, often in response to allergen hypersensi‑
esis holds true, this would suggest that ACOS should take tivity,19 the obstruction in COPD is thought to be incom‑

For personal use only 5 of 14


STAT E O F T H E A RT R E V I E W

pletely reversible and the product of environmental Air pollution


exposures such as tobacco or biomass fuel.75 Patients Ambient air pollution—a mixture of particulate matter,
with asthma who are exposed to noxious inhalants carbon monoxide, lead, sulphur dioxide, nitrogen oxides,
traditionally associated with COPD could conceivably and ozone—is a well known mediator of airways disease,
develop this degree of fixed obstruction over time. Sec‑ and multiple large population studies have shown that
ondly, in contrast to asthma, emphysema and the loss of long term exposure to air pollution is detrimental for lung
lung elastic recoil is an important phenotype of COPD.76 function. The Framingham Offspring or Third Generation
Autopsy studies, however, suggest that these processes studies, made up of more than 6000 participants, showed
may have been overlooked in asthma.77 78 Here, we review that proximity to a major roadway and exposure to fine
both aspects of COPD that may develop in patients with particulate matter (PM2.5) decreased FEV1 and acceler‑
asthma. ated decline in FEV1.100 Other components of air pollu‑
tion, such as nitrogen oxides and PM10, have also been
Smoking associated with low FEV1.101 Conversely, improvements in
A quarter of patients with asthma are current ciga‑ PM10 and PM2.5 levels over time increased FEV1 and FVC
rette smokers,79 and in comparison with non-smoking in children residing in southern California.102 For patients
patients with asthma, they carry a significantly greater with asthma in particular, exposure to excess air pollu‑
risk for developing fixed airflow obstruction and tion has considerable deleterious effects, with exacerba‑
ACOS.41‑82 Exposure to cigarette smoke in asthma seems tions, emergency room visits, and hospital admissions
to remodel the airway in a way that may worsen small all closely associated with levels of air pollution.103‑107
airways obstruction. Firstly, although goblet cell hyper‑ The effect of air pollution on the asthmatic airway has
plasia and mucus hypersecretion are well described multiple facets: impaired regulatory T cell function,108
pathological findings in both asthma and COPD, activation of toll-like receptor (TLR)-2 and TLR-4 medi‑
these features are aggravated in current smokers with ated innate immune responses,109 increased oxidative
asthma compared with ex-smokers or non-smokers with stress,110‑113 cytokine mediated airway inflammation,114 115
asthma.83 Secondly, multiple studies using both induced and alterations in DNA methylation.116 117
sputum samples and endobronchial biopsies have now Any or all of these mechanisms could conceivably
shown that smoking in asthma increases airway neu‑ induce the phenotypic switch from asthma to ACOS,
trophilia, shifting the predominantly eosinophilic cell although data confirming this hypothesis are notably
pattern typically observed in asthma towards a neutro‑ lacking. Nonetheless, a study investigating the develop‑
philic COPD pattern.84‑88 This process may be mediated ment of COPD in Ontario’s asthma population provides
by cytokines, as interleukin (IL)-6, IL-8, and IL-17A have the most convincing evidence to date for a link between
all been implicated in n­eutrophil chemotaxis in smokers air pollution and ACOS.8 In this study, 6040 adults in
with asthma.85‑89 In addition, smoking may induce CD8+ Ontario, Canada, with asthma were followed between
T cell proliferation in the asthmatic airway,90 similar to 1996 and 2014, with a diagnosis of COPD by a physician
COPD.91 92 Studies have shown that both neutrophil and during that time period used as the definition of ACOS. Air
CD8+ T cell numbers correlate well with increasing air‑ quality data were provided from 49 continuous fixed site
flow obstruction.85 93 Increases in CD8+ T cells in par‑ monitors throughout Ontario. For every increase in PM2.5
ticular may relate to faster decline in lung function,93 as of 10 μg/m3, the adjusted hazard ratio for developing
well as incomplete reversibility of airflow obstruction.94 ACOS was 2.78 (95% confidence interval 1.62 to 4.78).
Experimental models in which allergen sensitised mice Environmental exposures may thus be one important trig‑
are exposed to cigarette smoke seem to confirm these ger in the pathogenesis of ACOS.
findings, with attenuation of eosinophilic inflammation
in favour of increases in neutrophils, CD4+ and CD8+ T Loss of lung elastic recoil and development of
cells, and total cells.95‑98 emphysema in asthma
The recent application of new ’omics platforms to Emphysema, hyperinflation, and the loss of lung elastic
asthmatic smokers has shed light on novel genes and recoil are often associated with COPD, but they may also
proteins that may play a role in shifting asthma to ACOS, play an integral role in the later stages of severe asthma.
although the direct connection from these findings to Recognition of the loss of lung elastic recoil dates back
disease pathogenesis is still being clarified. Proteomic to 1967 when Gold, Kaufman, and Nadel showed that
analysis of induced sputum samples in four different patients with severe asthma lasting longer than five years
asthma phenotypes found that, compared with non- had significantly reduced lung elastic recoil compared
smoking patients with severe asthma, smokers and with healthy people.118 Initially, this was not thought to
ex-smokers with severe asthma had decreased sputum be related to emphysematous tissue destruction, as elas‑
abundance of tyrosine protein kinase Lyn (involved in tic recoil would return to normal following treatment with
mast cell degranulation).99 Genes up-regulated in the bronchodilators. However, subsequent studies showed
smoking group predominantly reflect actin cytoskeleton that the loss of lung elastic recoil was more durable than
function. Validation of these target proteins and genes anticipated and that this loss can still be observed in sta‑
in independent cohorts and deeper interrogation of the ble clinical states.119‑121 Normal diffusing capacity and
mechanistic pathways underlying their role in smoking computed tomography imaging again argued against the
related changes in the asthmatic airway may provide fur‑ presence of significant emphysema as a cause of elastic
ther insight into the origins of ACOS. recoil abnormalities.120

For personal use only 6 of 14


STAT E O F T H E A RT R E V I E W

Two autopsy studies have since shifted our understand‑ represent advanced airflow obstruction regardless of the
ing of the structural lung changes in asthma. A study in underlying airways disease).127 128 Airway wall thicken‑
2004 found a decrease in elastic fibres and an increase ing, airway smooth muscle (ASM) proliferation, and the
in the number of abnormal alveolar attachments in fatal obliteration of the terminal bronchioles develop over time
asthma compared with controls.78 A subsequent study in in COPD and, along with mucous secretions, contribute
2014 reported the autopsy findings of three patients with to this airway narrowing.129‑131 The degree of airway
asthma, which showed that although macroscopic evalu‑ hyper-responsiveness depends on the balance between
ation of the lungs did not show emphysema, microscopic ASM shortening and the opposite recoil force of the sur‑
examination showed diffuse upper lobe predominant rounding tissue, which in COPD may be compromised
mild centrilobular emphysema.122 More recently, it has by the development of emphysema and the destruction
been suggested that recurrent exacerbations of asthma of elastic fibres.132 As the airway wall thickens and as
induce bronchiolar inflammation, with activation of ASM area increases, a larger ASM contraction against a
proteases such as neutrophil elastase, cathepsin G, and weaker opposing elastic load allows for a larger degree
matrix metalloproteases (MMP), all of which conspire to of airway narrowing. Whether we can easily equate the
break down the lung parenchyma.77 In vitro work has process of airway hyper-responsiveness in COPD with
implicated IL-13 as a potent suppressor of elastin mRNA that in asthma is not clear, however. The contribution
expression through a pathway that involves the secretion of changes in the extracellular matrix to airway hyper-
of MMP-1 and MMP-2.123 The later stages of asthma may responsiveness is much larger in COPD than in asthma,
thus start to resemble emphysema, shifting asthma down whereas the airway hyper-responsiveness in asthma is
the airways spectrum to ACOS. driven primarily by ASM hypertrophy and hyperplasia.133
Therefore, although these processes may look similar on
How might COPD become ACOS? provocation testing, treatments targeting airway hyper-
The reverse scenario, in which a patient with COPD devel‑ responsiveness in asthma may not necessarily translate to
ops features of asthma, is admittedly less compelling. COPD patients who develop airway hyper-responsiveness
This would require the development of three important and evolve to ACOS.
pathological traits of asthma in a patient with COPD:
allergen sensitisation, airway hyper-responsiveness, and Airway inflammation
eosinophilic and type 2 (Th2) mediated airway inflam‑ Conventional understanding of COPD places emphasis
mation. Allergen sensitisation has been known to occur on a neutrophilic, type 1 (Th1) inflammatory airway
in COPD, particularly in older people,124 and upwards of response,134 135 but a subset of COPD patients who do
25-30% of COPD patients report allergic upper airway not cleanly fit this paradigm clearly exist. For instance,
symptoms or show IgE sensitisation to perennial aller‑ peripheral blood eosinophil concentrations persistently
gens.125 Among smokers, allergen sensitisation has also above 2% over a three year period were found in just
been associated with faster decline in lung function and, under 40% of participants in the ECLIPSE (Evaluation
among COPD patients, with increased respiratory symp‑ of COPD Longitudinally to Identify Predictive Surrogate
toms and COPD exacerbations.125 126 Whether allergen End-points) cohort.136 Recently, Christensen et al applied
sensitisation ultimately remodels the airway in the man‑ a 100 gene signature corresponding to high Th2 inflam‑
ner of asthma has yet to be demonstrated, however, and mation to airway epithelial cells from COPD patients.137
little research on this has been done. Similarly, airway Approximately 5% of smokers with COPD showed a high
hyper-responsiveness and eosinophilic/Th2 mediated Th2 gene expression signature, which was associated
inflammation also occur in COPD. However, for the for‑ with increased serum and tissue eosinophil counts,
mer it is by no means certain whether the pathological increased bronchodilator responsiveness, and greater
process is the same as in asthma, and for the latter it is improvement in hyperinflation after treatment with
conceivable that similar inflammatory changes also occur inhaled corticosteroids. This signature also notably cor‑
in patients with asthma who develop ACOS. Below, we related with the gene expression of mast cell marker CPA3
review the mechanisms by which these two features may and eosinophil chemotactic molecule CCL26. Whether
emerge or worsen over the course of COPD. this signature has identified a unique endotype of COPD
or has isolated an important ACOS airway epithelial gene
Airway hyper-responsiveness and airway remodelling signature is unclear; the authors recommend that the sig‑
Airway hyper-responsiveness is the acute response of an nature be studied in cohorts better phenotyped for both
airway to a bronchial challenge, often in the form of an these conditions.
agent such as methacholine or histamine. Airway hyper- Despite the concerns about the quality of phenotyping
responsiveness affected up to a quarter of participants in Christensen et al’s cohorts, other authors investigating
in the Lung Health Study and was associated with faster sputum cytokine clusters would concur with their hypoth‑
decline in FEV1 and higher respiratory related mortality.48 esis that an overlap phenotype of eosinophilic COPD can
The response to a bronchial challenge is approximately be readily recognised.138 In a cohort of patients with
25-50% more robust in moderate to severe COPD than severe asthma and COPD, three clusters were identified
in mild COPD. This might suggest that this later stage on the basis of sputum cytokine measurements: asthma
development may simply be geometric in nature, as the with high Th2 and eosinophilic inflammation, asthma
resistance of the airway is inversely proportional to the and COPD with sputum neutrophil predominance, and
radius to the fourth power (and therefore may merely COPD with predominantly eosinophilic inflammation.

For personal use only 7 of 14


STAT E O F T H E A RT R E V I E W

This latter group, corresponding to the high Th2 COPD physiology and even its definition, implies that no single
group in Christenson et al’s study, featured increased con‑ class of treatment is likely to help all patients. Impor‑
centrations of important Th2 cytokines and chemokines— tantly, we and others have advocated that the field move
namely, IL-6, CCL2, CCL13, and CCL17. Although these towards deeper phenotyping of these patients to guide
studies have been instrumental in exposing the sig‑ treatment. In particular, as proposed by Barnes, three
nificance of Th2 responses in COPD, why certain COPD phenotypes of ACOS have been described that are impor‑
patients are driven towards this response compared with tant to establish before starting treatment: eosinophilic
others who remain in the Th1, neutrophilic realm is still COPD (high Th2 inflammation), neutrophilic asthma
unclear. (low Th2 inflammation), and paucigranulocytic ACOS.142
Needless to say, improved access to and standardisation
Is ACOS its own disease? of sputum cytology measurements is needed before such
Finally, in accordance with the British hypothesis, we a strategy can be implemented. Even so, this framework
consider a third scenario in which ACOS has a unique provides a useful algorithm for targeting treatment of
origin unrelated to either asthma or COPD. Some of the ACOS, one rooted in the presumed pathophysiology of
strongest evidence against the Dutch hypothesis has been the disease. In this section, we present the evidence for
the striking lack of a unifying genetic cause of asthma therapeutic options based on these three phenotypes,
and COPD despite many genome-wide association stud‑ with the caveat that few trials have specifically investi‑
ies (GWAS).139 Whether ACOS will follow this pattern and gated patients with ACOS. Extrapolations from asthma
show its own genetic determinants separate from those and COPD trials are thus also presented.
of asthma or COPD has yet to be definitively answered.
One GWAS comparing patients with COPD and ACOS in Eosinophilic COPD
the COPDGene Study did not find any single nucleotide Inhaled corticosteroids
polymorphisms (SNPs) associated with ACOS that met the Targeted against eosinophilic pathology, inhaled corti‑
pre-determined genome-wide significance threshold of costeroids and combinations with long acting β agonists
5×10−8, although the authors noted that the study was (LABA) have naturally featured prominently in treat‑
probably underpowered to detect this degree of differ‑ ment recommendations for ACOS given the emphasis
ence.16 SNPs that approached genome-wide significance many groups have placed on eosinophilia as a diagnos‑
included those on the genes CSMD1 and GPR65, the for‑ tic criterion.143 144 In addition, inhaled corticosteroid/
mer a tumour suppressor gene previously implicated in LABA combinations are recommended in both asthma
emphysema and the latter a mediator of eosinophil acti‑ and COPD, making their use in ACOS appealing. The
vation in asthma.140 141 Clearly, genetic work on ACOS strength of evidence for these recommendations is mixed,
remains preliminary and larger studies are needed to however. Trials evaluating inhaled corticosteroids alone
better establish its genetic determinants. and in combination with LABA have been small in size
If asthma and COPD are not in fact prerequisite stages and mostly retrospective and observational in nature.
to be passed through before developing ACOS, it is con‑ Moreover, results have been conflicting. A 12 year obser‑
ceivable that events or insults in early childhood might vational retrospective cohort study in which 90 patients
trigger the shift towards asthma, COPD, or ACOS, each with ACOS treated with inhaled corticosteroids were com‑
having its own distinct trajectory. In support of this pared with 35 ACOS patients not treated with inhaled
hypothesis, the Tasmanian Longitudinal Health Study corticosteroids found no difference in decline in FEV1,
analysed lung function collected at age 7 and then at age exacerbation rates, or overall mortality.145 On the other
45, determining whether childhood spirometry could hand, a prospective study found that 127 ACOS patients
predict the development of ACOS in adulthood (defined who received inhaled budesonide had improved spirom‑
as a self report of asthma in conjunction with a post-bron‑ etry and measures of hyperinflation, sputum eosinophils,
chodilator FEV1/FVC below the lower limit of normal).46 serum IgE, and FeNO after treatment.146 The lack of a true
Those in the lowest quarters of FEV1/FVC and FEV1 per placebo group in this trial, however, limits the generalis‑
cent predicted at age 7 had a 16.3 and 2.93 greater odds, ability of these results. Compared with COPD patients,
respectively, of developing ACOS by age 45. This associa‑ those with ACOS may show better FEV1 response to
tion, which persisted even after accounting for smoking, inhaled corticosteroid/LABA treatments, but again, these
was far greater than that between childhood lung func‑ trials featured no more than 45 ACOS patients.52 147 The
tion and COPD, whereas no association with asthma was possibility that inhaled corticosteroids with or without
noted. Whether due to in utero and/or childhood expo‑ LABA may provide benefit for eosinophilic ACOS patients
sure to smoke or childhood respiratory infections, the cannot be either discounted or definitively proven given
inability to reach maximum lung function during child‑ the amount and quality of data at hand.
hood, already discernible by age 7, seems to have lasting
implications for future airways disease. Anti-IgE therapies
Severe, persistent asthma that remains uncontrolled
Possibilities for therapeutic intervention despite inhaled corticosteroids and inhaled corticoster‑
The myriad ways in which a particular person can poten‑ oid/LABA combinations now prompts consideration of
tially develop ACOS poses considerable challenges for anti-IgE treatments, including recombinant humanised
treatment. The very heterogeneity of ACOS, the untoward monoclonal antibodies such as omalizumab. The binding
by-product of a lack of consensus surrounding its patho‑ of IgE to the IgE receptor FcεR1 on basophils and mast

For personal use only 8 of 14


STAT E O F T H E A RT R E V I E W

cells triggers cytokine mediated eosinophilic inflam‑ seem to show greater efficacy against asthma than do the
mation; blockage of this linkage by compounds such as IL-13 agents, both have yet to be tested in eosinophilic
omalizumab helps to attenuate this cascade, which ulti‑ COPD, so extrapolation to ACOS is merely speculative at
mately has been shown to reduce exacerbations, hospital this time.
admissions, emergency department visits, and inhaled
corticosteroid and rescue inhaler use in patients with Neutrophilic asthma—macrolides
advanced asthma.148 Recently, the efficacy of omalizumab The pleiotropic properties of macrolides, functioning
was assessed in patients with severe allergic asthma at once as antibacterial, immunomodulatory, and anti-
enrolled in the Australian Xolair Registry who also had inflammatory drugs, have resulted in their wide appli‑
a diagnosis of COPD, whether by physician assessment cation in several neutrophilic respiratory disorders such
(n=17) or by fixed airflow obstruction on spirometry as cystic fibrosis and non-cystic fibrosis bronchiectasis.
(n=55).149 Regardless of the definition of COPD, these Macrolides seem to dampen IL-8 and CXCL1, leading to
ACOS patients showed significantly improved asthma neutrophil apoptosis and decreased oxidative stress.164
control and health related quality of life scores (exceed‑ Importantly, azithromycin has become a therapeutic
ing the minimally clinically important difference) follow‑ option in COPD, in which its long term use was shown to
ing treatment with omalizumab despite no significant reduce the frequency of exacerbations,165 and whether
improvements in their FEV1. These incremental gains these effects could translate to neutrophilic forms of
were equivalent to those observed in patients with asthma asthma has been the subject of several studies. To date,
alone. Smaller case series of three and 10 ACOS patients the results have been modest at best. Although the larger
treated with omalizumab each echoed these improve‑ randomised controlled trials focusing on patients with
ments in asthma symptom control.150 151 non-eosinophilic asthma have shown a reduction in exac‑
erbation rates and duration of acute events with long term
Anti-IL-5/IL-5Rα therapies macrolide use,166 167 other trials have shown no significant
IL-5 plays a key role in eosinophil differentiation and benefit on either these outcomes or asthma symptom con‑
maturation in the bone marrow, as well as migration trol and lung function.168‑174 The 2015 Cochrane review
from the blood to tissue sites.152 Three new anti-IL-5 of macrolides in chronic asthma concluded that, on the
and anti-IL-5Rα monoclonal antibodies (mepolizumab, basis of very low quality evidence, macrolides had no dis‑
benralizumab, and reslizumab) have now been studied cernible benefit in reducing exacerbations or in improv‑
in eosinophilic asthma with impressive improvements ing asthma control, quality of life, and rescue medication
in exacerbation rates, asthma control scores, and ster‑ use.175 A small positive effect on lung function could be
oid usage.153‑156 These compounds have yet to be tested observed, suggesting that there may still be some clinical
in cohorts of ACOS patients, but a randomised, double benefit to certain asthma patients; however, large effect
blind, placebo controlled study evaluating benralizumab sizes for clinically important endpoints in asthma are
in COPD patients with sputum eosinophil counts above unlikely to be achieved with macrolides.
3% was far from promising.157 Benralizumab failed to
reduce the exacerbation rate, the primary endpoint, Paucigranulocytic ACOS—long acting muscarinic
although improvements in spirometry seemed to occur. antagonists
For the subgroup of patients with baseline blood eosino‑ For those ACOS patients without a distinct cellular inflam‑
phil counts above 200 cells/μL, numerical but non-sig‑ matory response, we return to bronchodilators, the stal‑
nificant improvements in exacerbation rates, FEV1, and wart of airways disease treatment. Ideally, for the patient
St George’s Respiratory and Chronic Respiratory Disease with ACOS, bronchodilators should reduce ASM tone and
Questionnaire scores occurred, but until further large dynamic hyperinflation. We have described the role of
randomised controlled trials are conducted, the role inhaled corticosteroid/LABA combinations in ACOS
for anti-IL-5 and anti-IL-5Rα monoclonal antibodies in above; here we discuss whether long acting muscarinic
eosinophilic COPD is unclear. antagonists (LAMA) widely used in COPD have any benefit
in asthma and, by extension, ACOS. Studies assessing the
Anti-IL-13/IL-4Rα therapies efficacy of LAMA therapy in asthma have largely evalu‑
IL-13 and IL-4 are important Th2 cytokines that contrib‑ ated tiotropium, an M1 and M3 receptor antagonist with
ute to several asthma related pathologies including the a proven benefit in COPD. Several large, high quality ran‑
regulation of eosinophils, mucus production, goblet cell domised controlled trials (one of which enrolled asthma
hyperplasia, ASM contraction, and airway remodelling.148 patients with persistent post-bronchodilator airflow limi‑
Anti-IL-13 antibodies such as lebrikizumab and traloki‑ tation176) have consistently shown improvements in FEV1
numab target IL-13, whereas anti-IL-4Rα antibodies such in patients with asthma, both in children and in adults,
as dupilumab target both IL-13 and IL-4. In patients with when tiotropium is added to inhaled corticosteroid and
severe, persistent asthma, these monoclonal antibodies inhaled corticosteroid/LABA therapies.176‑182 Several stud‑
have shown improvements in lung function and exacer‑ ies have also shown a reduction in exacerbation risk,179 183
bation rates.158‑162 The efficacy of these agents seems to be as well as asthma symptom control.179‑182 Whether LAMA
enhanced in patients with high serum concentrations of compounds other than tiotropium have similar positive
periostin, an extracellular matrix protein deposited in the effects is yet to be tested, but the clinical improvements
basement membrane of asthmatic airways that reflects observed in both COPD and asthma patients suggest that
Th2 inflammation.163 Although the IL-4Rα antagonists LAMA therapies may be an option for patients with ACOS.

For personal use only 9 of 14


STAT E O F T H E A RT R E V I E W

GLOSSARY OF ABBREVIATIONS QUESTIONS FOR FUTURE RESEARCH


ACOS—asthma-COPD overlap syndrome • Do unique genetic, molecular, and histological patterns
ASM—airway smooth muscle exist that distinguish asthma-COPD overlap syndrome
COPD—chronic obstructive pulmonary disease (ACOS) from asthma or chronic obstructive pulmonary
disease (COPD) alone?
FeNO—fractional exhaled nitric oxide
• What are the long term clinical outcomes for patients with
FEV1—forced expiratory volume in 1 s
ACOS, and how might these outcomes vary depending on
FVC—forced vital capacity
the definition used for ACOS?
GINA—Global Initiative for Asthma
• Should first line treatment for patients with ACOS include
GOLD—Global Initiative for Chronic Obstructive Lung inhalers containing inhaled corticosteroid?
Disease
• Do patients with ACOS benefit from standard treatments
GWAS—genome-wide association studies for asthma and COPD, or are novel compounds needed
IgE—immunoglobulin E specifically tailored to the ACOS phenotype?
IL—interleukin
LABA—long acting β agonist
clinician in an otherwise data-free zone. Greater empha‑
LAMA—long acting muscarinic antagonist
sis on the ACOS population in future research is recom‑
MMP—matrix metalloprotease
mended.
SNPs—single nucleotide polymorphism
We thank Cheng Wei Tony Yang and Chloe Chih Ya Huang for their
contribution to the figures.
Emerging treatments Contributors: Both authors were involved in the conception, writing, and
editing of the manuscript. Both are guarantors.
Although several ongoing studies are aiming to charac‑
Competing interests: We have read and understood BMJ policy on
terise ACOS further, no clinical trials are being conducted declaration of interests and declare the following interests: DDS has
to assess the utility of either novel or known treatments received honorariums for speaking engagements with AstraZeneca and
in ACOS patients. Furthermore, attempts to apply avail‑ Boehringer Ingelheim and for participating in COPD advisory boards of
Sanofi, Regeneron, AstraZeneca, Boehringer Ingelheim, Novartis, and
able asthma therapies to COPD patients with an eosino‑ Merck and has received research funding for investigator initiated COPD
philic phenotype have garnered mixed results. One phase studies from Merck, AstraZeneca, and Boehringer Ingelheim. DDS is
III trial evaluating mepolizumab in eosinophilic COPD supported by a Tier 1 Canada research chair award in COPD.
showed a statistically significant reduction in moderate Provenance and peer review: Commissioned; externally peer reviewed.
to severe exacerbations compared with placebo, whereas Patient involvement: No patients were involved in the creation of this article.
another failed to show any difference.184 Whether future 1 Orie NG, Sluiter HJ. Bronchitis.Charles C Thomas, 1961.
2 Barnes PJ. Against the Dutch hypothesis: asthma and chronic
trials will be performed to resolve these conflicting results obstructive pulmonary disease are distinct diseases. Am J Respir
has yet to be announced. Crit Care Med 2006;174:240-3, discussion 243-4. doi:10.1164/
rccm.2604008 pmid:16864717.
3 Kraft M. Asthma and chronic obstructive pulmonary disease exhibit
Guidelines common origins in any country!Am J Respir Crit Care Med 2006;174:238-
40, discussion 243-4. doi:10.1164/rccm.2604007 pmid:16864716.
To date, no guideline statements grading the quality of
4 Vermeire PA, Pride NBA. A “splitting” look at chronic nonspecific lung
evidence surrounding ACOS have been issued. Expert disease (CNSLD): common features but diverse pathogenesis. Eur Respir J
panel consensus statements (fig 1), however, have been 1991;4:490-6.pmid:1855578.
5 Alshabanat A, Zafari Z, Albanyan O, Dairi M, FitzGerald JM. Asthma
published with general agreement about two principles: and COPD Overlap Syndrome (ACOS): A Systematic Review and Meta
that ACOS should be considered in any patient over the Analysis. PLoS One 2015;10:e0136065. doi:10.1371/journal.
pone.0136065 pmid:26336076.
age of 40 with persistent airflow obstruction and a history 6 Goto T, Camargo CA Jr, , Hasegawa K. Fractional exhaled nitric oxide levels
of asthma or evidence of bronchodilator reversibility and in asthma-COPD overlap syndrome: analysis of the National Health and
Nutrition Examination Survey, 2007-2012. Int J Chron Obstruct Pulmon
that the treatment of ACOS should include inhaled corti‑ Dis 2016;11:2149-55. doi:10.2147/COPD.S110879 pmid:27660432.
costeroids. However, these statements acknowledge the 7 Ding B, DiBonaventura M, Karlsson N, Ling X. Asthma-chronic obstructive
pulmonary disease overlap syndrome in the urban Chinese population:
need for additional evidence for these recommendations. prevalence and disease burden using the 2010, 2012, and 2013 China
National Health and Wellness Surveys. Int J Chron Obstruct Pulmon Dis
Conclusions 2016;11:1139-50.pmid:27354777.
8 To T, Zhu J, Larsen K, et al. Canadian Respiratory Research Network.
ACOS may represent a distinct clinical phenotype of Progression from Asthma to Chronic Obstructive Pulmonary Disease. Is
asthma and COPD and, importantly for clinicians, is Air Pollution a Risk Factor?Am J Respir Crit Care Med 2016;194:429-38.
doi:10.1164/rccm.201510-1932OC pmid:26950751.
marked by worse respiratory symptoms and rates of 9 Woodruff PG, van den Berge M, Boucher RC, et al. American Thoracic
exacerbation and hospital admission. For these rea‑ Society/National Heart, Lung, and Blood Institute Asthma-Chronic
Obstructive Pulmonary Disease Overlap Workshop Report. Am J Respir
sons, the diagnosis of ACOS should be considered in all Crit Care Med 2017;196:375-81. doi:10.1164/rccm.201705-
patients over the age of 40 years with persistent airflow 0973WS pmid:28636425.
10 Kurashima K, Takaku Y, Ohta C, Takayanagi N, Yanagisawa T, Sugita Y.
obstruction, a history of cigarette smoking or exposure to COPD assessment test and severity of airflow limitation in patients
biomass fuel, and a history of asthma or strong broncho‑ with asthma, COPD, and asthma-COPD overlap syndrome. Int J
dilator reversibility on spirometry. Although the pathway Chron Obstruct Pulmon Dis 2016;11:479-87. doi:10.2147/COPD.
S97343 pmid:27019598.
to ACOS has yet to be elucidated in its entirety, one can 11 Kauppi P, Kupiainen H, Lindqvist A, et al. Overlap syndrome of asthma
speculate that several ways exist in which a patient with and COPD predicts low quality of life. J Asthma 2011;48:279-85.
doi:10.3109/02770903.2011.555576 pmid:21323613.
asthma can develop a neutrophilic “COPD-like” airway 12 de Marco R, Pesce G, Marcon A, et al. The coexistence of asthma and
and a COPD patient can develop an eosinophilic, reac‑ chronic obstructive pulmonary disease (COPD): prevalence and risk
factors in young, middle-aged and elderly people from the general
tive “asthma-like” airway. Basing therapeutic choices on population. PLoS One 2013;8:e62985. doi:10.1371/journal.
these inflammatory distinctions may help to guide the pone.0062985 pmid:23675448.

For personal use only 10 of 14


STAT E O F T H E A RT R E V I E W

13 Andersén H, Lampela P, Nevanlinna A, Säynäjäkangas O, Keistinen T. High 36 Rossall MR, Cadden PA, Molphy SD, Plumb J, Singh D. Repeatability


hospital burden in overlap syndrome of asthma and COPD. Clin Respir J of induced sputum measurements in moderate to severe
2013;7:342-6. doi:10.1111/crj.12013 pmid:23362945. asthma. Respir Med 2014;108:1566-8. doi:10.1016/j.
14 Bai JW, Mao B, Yang WL, Liang S, Lu HW, Xu JF. Asthma-COPD rmed.2014.08.004 pmid:25195138.
overlap syndrome showed more exacerbations however lower 37 Petsky HL, Cates CJ, Lasserson TJ, et al. A systematic review and meta-
mortality than COPD. QJM 2017;pii:hcx005. doi:10.1093/qjmed/ analysis: tailoring asthma treatment on eosinophilic markers (exhaled
hcx005 pmid:28100824. nitric oxide or sputum eosinophils). Thorax 2012;67:199-208.
15 Kim MA, Noh CS, Chang YJ, et al. Asthma and COPD overlap syndrome is doi:10.1136/thx.2010.135574 pmid:20937641.
associated with increased risk of hospitalisation. Int J Tuberc Lung Dis 38 Gochicoa-Rangel L, Rojas-Cisneros F, Miguel-Reyes JL, et al. Variability
2015;19:864-9. doi:10.5588/ijtld.14.0327 pmid:26056115. of FeNO in healthy subjects at 2240 meters above sea level. J Clin
16 Hardin M, Cho M, McDonald ML, et al. The clinical and genetic features Monit Comput 2016;30:445-9. doi:10.1007/s10877-015-9737-
of COPD-asthma overlap syndrome. Eur Respir J 2014;44:341-50. 0 pmid:26174797.
doi:10.1183/09031936.00216013 pmid:24876173. 39 Kapande KM, McConaghy LA, Douglas I, et al. Comparative repeatability of
17 Sin DD, Miravitlles M, Mannino DM, et al. What is asthma-COPD overlap two handheld fractional exhaled nitric oxide monitors. Pediatr Pulmonol
syndrome? Towards a consensus definition from a round table discussion. 2012;47:546-50. doi:10.1002/ppul.21591 pmid:22038831.
Eur Respir J 2016;48:664-73. doi:10.1183/13993003.00436- 40 de Marco R, Marcon A, Rossi A, et al. Asthma, COPD and overlap syndrome:
2016 pmid:27338195. a longitudinal study in young European adults. Eur Respir J 2015;46:671-
18 Gibson PG, Simpson JL. The overlap syndrome of asthma and COPD: 9. doi:10.1183/09031936.00008615 pmid:26113674.
what are its features and how important is it?Thorax 2009;64:728-35. 41 Chung JW, Kong KA, Lee JH, Lee SJ, Ryu YJ, Chang JH. Characteristics and
doi:10.1136/thx.2008.108027 pmid:19638566. self-rated health of overlap syndrome. Int J Chron Obstruct Pulmon Dis
19 Global Initiative for Asthma. Global strategy for asthma management and 2014;9:795-804.pmid:25092973.
prevention. 2015 http://ginasthma.org/wp-content/uploads/2016/01/ 42 Kumbhare S, Pleasants R, Ohar JA, Strange C. Characteristics and
GINA_Report_2015_Aug11-1.pdf. Prevalence of Asthma/Chronic Obstructive Pulmonary Disease Overlap in
20 Soler-Cataluña JJ, Cosío B, Izquierdo JL, et al. Consensus document on the United States. Ann Am Thorac Soc 2016;13:803-10. doi:10.1513/
the overlap phenotype COPD-asthma in COPD. Arch Bronconeumol AnnalsATS.201508-554OC pmid:26974689.
2012;48:331-7. doi:10.1016/j.arbr.2012.06.017 pmid:22341911. 43 Menezes AMB, Montes de Oca M, Pérez-Padilla R, et al. PLATINO Team.
21 Cosio BG, Soriano JB, López-Campos JL, et al. CHAIN Study. Defining the Increased risk of exacerbation and hospitalization in subjects with
Asthma-COPD Overlap Syndrome in a COPD Cohort. Chest 2016;149:45- an overlap phenotype: COPD-asthma. Chest 2014;145:297-304.
52. doi:10.1378/chest.15-1055 pmid:26291753. doi:10.1378/chest.13-0622 pmid:24114498.
22 Koblizek V, Chlumsky J, Zindr V, et al. Czech Pneumological and 44 Pleasants RA, Ohar JA, Croft JB, et al. Chronic obstructive pulmonary
Phthisiological Society. Chronic Obstructive Pulmonary Disease: official disease and asthma-patient characteristics and health impairment. COPD
diagnosis and treatment guidelines of the Czech Pneumological and 2014;11:256-66.pmid:24152212.
Phthisiological Society; a novel phenotypic approach to COPD with 45 Diaz-Guzman E, Khosravi M, Mannino DM. Asthma,
patient-oriented care. Biomed Pap Med Fac Univ Palacky Olomouc Czech chronic obstructive pulmonary disease, and mortality
Repub 2013;157:189-201.pmid:23733084. in the U.S. population. COPD 2011;8:400-7.
23 Cataldo D, Corhay JL, Derom E, et al. A Belgian survey on the diagnosis doi:10.3109/15412555.2011.611200 pmid:22149399.
of asthma-COPD overlap syndrome. Int J Chron Obstruct Pulmon Dis 46 Bui DS, Burgess JA, Lowe AJ, et al. Childhood Lung Function Predicts
2017;12:601-13. doi:10.2147/COPD.S124459 pmid:28243078. Adult Chronic Obstructive Pulmonary Disease and Asthma-Chronic
24 Jin J, Liu X, Sun Y. The prevalence of increased serum IgE and Aspergillus Obstructive Pulmonary Disease Overlap Syndrome. Am J Respir
sensitization in patients with COPD and their association with symptoms Crit Care Med 2017;196:39-46. doi:10.1164/rccm.201606-
and lung function. Respir Res 2014;15:130. doi:10.1186/s12931-014- 1272OC pmid:28146643.
0130-1 pmid:25359094. 47 Wheaton AG, Pleasants RA, Croft JB, et al. Gender and asthma-chronic
25 Tamada T, Sugiura H, Takahashi T, et al. Biomarker-based detection obstructive pulmonary disease overlap syndrome. J Asthma 2016;53:720-
of asthma-COPD overlap syndrome in COPD populations. Int J Chron 31. doi:10.3109/02770903.2016.1154072 pmid:27043854.
Obstruct Pulmon Dis 2015;10:2169-76. doi:10.2147/COPD. 48 Tkacova R, Dai DL, Vonk JM, et al. Airway hyperresponsiveness in
S88274 pmid:26491283. chronic obstructive pulmonary disease: A marker of asthma-chronic
26 Bafadhel M, McKenna S, Terry S, et al. Blood eosinophils to direct obstructive pulmonary disease overlap syndrome?J Allergy Clin Immunol
corticosteroid treatment of exacerbations of chronic obstructive 2016;138:1571-9. doi:10.1016/j.jaci.2016.04.022 pmid:27345171.
pulmonary disease: a randomized placebo-controlled trial. Am J 49 Baarnes CB, Kjeldgaard P, Nielsen M, Miravitlles M, Ulrik CS. Identifying
Respir Crit Care Med 2012;186:48-55. doi:10.1164/rccm.201108- possible asthma-COPD overlap syndrome in patients with a new diagnosis
1553OC pmid:22447964. of COPD in primary care. NPJ Prim Care Respir Med 2017;27:16084.
27 Oshagbemi OA, Burden AM, Braeken DC, et al. Stability of Blood doi:10.1038/npjpcrm.2016.84 pmid:28055002.
Eosinophils in Patients with Chronic Obstructive Pulmonary Disease and 50 Caillaud D, Chanez P, Escamilla R, et al. Initiatives BPCO scientific
in Control Subjects, and the Impact of Sex, Age, Smoking, and Baseline committee and investigators. Asthma-COPD overlap syndrome (ACOS)
Count. Am J Respir Crit Care Med 2017;195:1402-4. doi:10.1164/ vs ‘pure’ COPD: a distinct phenotype?Allergy 2017;72:137-45.
rccm.201701-0009LE pmid:28165763. doi:10.1111/all.13004 pmid:27501862.
28 Couillard S, Larivée P, Courteau J, Vanasse A. Eosinophils 51 Miravitlles M, Huerta A, Fernández-Villar JA, et al. Generic utilities in chronic
in COPD Exacerbations Are Associated With Increased obstructive pulmonary disease patients stratified according to different
Readmissions. Chest 2017;151:366-73. doi:10.1016/j. staging systems. Health Qual Life Outcomes 2014;12:120. doi:10.1186/
chest.2016.10.003 pmid:27746201. s12955-014-0120-5 pmid:25189786.
29 Pavord ID, Lettis S, Anzueto A, Barnes N. Blood eosinophil count and 52 Kitaguchi Y, Komatsu Y, Fujimoto K, Hanaoka M, Kubo K. Sputum
pneumonia risk in patients with chronic obstructive pulmonary disease: eosinophilia can predict responsiveness to inhaled corticosteroid
a patient-level meta-analysis. Lancet Respir Med 2016;4:731-41. treatment in patients with overlap syndrome of COPD and asthma. Int J
doi:10.1016/S2213-2600(16)30148-5 pmid:27460163. Chron Obstruct Pulmon Dis 2012;7:283-9.pmid:22589579.
30 Watz H, Tetzlaff K, Wouters EF, et al. Blood eosinophil count and 53 Hardin M, Silverman EK, Barr RG, et al. COPDGene Investigators. The
exacerbations in severe chronic obstructive pulmonary disease after clinical features of the overlap between COPD and asthma. Respir Res
withdrawal of inhaled corticosteroids: a post-hoc analysis of the 2011;12:127. doi:10.1186/1465-9921-12-127 pmid:21951550.
WISDOM trial. Lancet Respir Med 2016;4:390-8. doi:10.1016/S2213- 54 Marsh SE, Travers J, Weatherall M, et al. Proportional classifications
2600(16)00100-4 pmid:27066739. of COPD phenotypes. Thorax 2008;63:761-7. doi:10.1136/
31 Pavord ID, Lettis S, Locantore N, et al. Blood eosinophils thx.2007.089193 pmid:18728201.
and inhaled corticosteroid/long-acting β-2 agonist 55 Sorino C, Pedone C, Scichilone N. Fifteen-year mortality of patients
efficacy in COPD. Thorax 2016;71:118-25. doi:10.1136/ with asthma-COPD overlap syndrome. Eur J Intern Med 2016;34:72-7.
thoraxjnl-2015-207021 pmid:26585525. doi:10.1016/j.ejim.2016.06.020 pmid:27357368.
32 Hasegawa K, Camargo CA Jr. Prevalence of blood eosinophilia in 56 Wurst KE, Rheault TR, Edwards L, Tal-Singer R, Agusti A, Vestbo J. A
hospitalized patients with acute exacerbation of COPD. Respirology comparison of COPD patients with and without ACOS in the ECLIPSE
2016;21:761-4. doi:10.1111/resp.12724 pmid:26699685. study. Eur Respir J 2016;47:1559-62. doi:10.1183/13993003.02045-
33 Simpson JL, McElduff P, Gibson PG. Assessment and reproducibility 2015 pmid:26989103.
of non-eosinophilic asthma using induced sputum. Respiration 57 Kobayashi S, Hanagama M, Yamanda S, Ishida M, Yanai M.
2010;79:147-51. doi:10.1159/000245899 pmid:19816004. Inflammatory biomarkers in asthma-COPD overlap syndrome. Int J
34 Akamatsu K, Matsunaga K, Sugiura H, et al. Improvement of Chron Obstruct Pulmon Dis 2016;11:2117-23. doi:10.2147/COPD.
Airflow Limitation by Fluticasone Propionate/Salmeterol S113647 pmid:27660429.
in Chronic Obstructive Pulmonary Disease: What is the 58 Chung WS, Lin CL, Kao CH. Comparison of acute respiratory events
Specific Marker?Front Pharmacol 2011;2:36. doi:10.3389/ between asthma-COPD overlap syndrome and COPD patients: a
fphar.2011.00036 pmid:21811461. population-based cohort study. Medicine (Baltimore) 2015;94:e755.
35 Chen FJ, Huang XY, Liu YL, Lin GP, Xie CM. Importance of fractional exhaled doi:10.1097/MD.0000000000000755 pmid:25929911.
nitric oxide in the differentiation of asthma-COPD overlap syndrome, 59 Milanese M, Di Marco F, Corsico AG, et al. ELSA Study Group. Asthma
asthma, and COPD. Int J Chron Obstruct Pulmon Dis 2016;11:2385-90. control in elderly asthmatics. An Italian observational study. Respir Med
doi:10.2147/COPD.S115378 pmid:27713629. 2014;108:1091-9. doi:10.1016/j.rmed.2014.05.016 pmid:24958604.

For personal use only 11 of 14


STAT E O F T H E A RT R E V I E W

60 Ozkaya S, Dirican A, Tuna T. The objective evaluation of obstructive 82 Kiljander T, Helin T, Venho K, Jaakkola A, Lehtimäki L. Prevalence of


pulmonary diseases with spirometry. Int J Chron Obstruct Pulmon Dis asthma-COPD overlap syndrome among primary care asthmatics with
2016;11:2009-15. doi:10.2147/COPD.S113774 pmid:27616884. a smoking history: a cross-sectional study. NPJ Prim Care Respir Med
61 Vaz Fragoso CA, Murphy TE, Agogo GO, Allore HG, McAvay GJ. Asthma- 2015;25:15047. doi:10.1038/npjpcrm.2015.47 pmid:26182124.
COPD overlap syndrome in the US: a prospective population-based 83 Broekema M, ten Hacken NH, Volbeda F, et al. Airway epithelial
analysis of patient-reported outcomes and health care utilization. Int changes in smokers but not in ex-smokers with asthma. Am J Respir
J Chron Obstruct Pulmon Dis 2017;12:517-27. doi:10.2147/COPD. Crit Care Med 2009;180:1170-8. doi:10.1164/rccm.200906-
S121223 pmid:28223792. 0828OC pmid:19797761.
62 Fu JJ, Gibson PG, Simpson JL, McDonald VM. Longitudinal changes 84 Boulet LP, Lemière C, Archambault F, Carrier G, Descary MC, Deschesnes F.
in clinical outcomes in older patients with asthma, COPD and Smoking and asthma: clinical and radiologic features, lung function,
asthma-COPD overlap syndrome. Respiration 2014;87:63-74. and airway inflammation. Chest 2006;129:661-8. doi:10.1378/
doi:10.1159/000352053 pmid:24029561. chest.129.3.661 pmid:16537865.
63 van Boven JF, Román-Rodríguez M, Palmer JF, Toledo- 85 Chalmers GW, MacLeod KJ, Thomson L, Little SA, McSharry C, Thomson NC.
Pons N, Cosío BG, Soriano JB. Comorbidome, Pattern, and Impact of Smoking and airway inflammation in patients with mild asthma. Chest
Asthma-COPD Overlap Syndrome in Real Life. Chest 2016;149:1011-20. 2001;120:1917-22. doi:10.1378/chest.120.6.1917 pmid:11742922.
doi:10.1016/j.chest.2015.12.002 pmid:26836892. 86 Siew LQC, Wu SY, Ying S, Corrigan CJ. Cigarette smoking increases
64 Cosentino J, Zhao H, Hardin M, et al. COPDGene Investigators. Analysis bronchial mucosal IL-17A expression in asthmatics, which acts in
of Asthma-Chronic Obstructive Pulmonary Disease Overlap Syndrome concert with environmental aeroallergens to engender neutrophilic
Defined on the Basis of Bronchodilator Response and Degree of inflammation. Clin Exp Allergy 2017;47:740-50. doi:10.1111/
Emphysema. Ann Am Thorac Soc 2016;13:1483-9. doi:10.1513/ cea.12907 pmid:28211191.
AnnalsATS.201511-761OC pmid:27268723. 87 St-Laurent J, Bergeron C, Pagé N, Couture C, Laviolette M, Boulet LP.
65 Nielsen M, Bårnes CB, Ulrik CS. Clinical characteristics of the asthma- Influence of smoking on airway inflammation and remodelling in
COPD overlap syndrome--a systematic review. Int J Chron Obstruct asthma. Clin Exp Allergy 2008;38:1582-9. doi:10.1111/j.1365-
Pulmon Dis 2015;10:1443-54.pmid:26251584. 2222.2008.03032.x pmid:18681853.
66 Kitaguchi Y, Yasuo M, Hanaoka M. Comparison of pulmonary function 88 Shimoda T, Obase Y, Kishikawa R, Iwanaga T. Influence of cigarette
in patients with COPD, asthma-COPD overlap syndrome, and asthma smoking on airway inflammation and inhaled corticosteroid treatment in
with airflow limitation. Int J Chron Obstruct Pulmon Dis 2016;11:991-7. patients with asthma. Allergy Asthma Proc 2016;37:50-8. doi:10.2500/
doi:10.2147/COPD.S105988 pmid:27274220. aap.2016.37.3944 pmid:27401308.
67 Suzuki T, Tada Y, Kawata N, et al. Clinical, physiological, and 89 Huang CC, Wang CH, Fu CH, et al. Association between cigarette smoking
radiological features of asthma-chronic obstructive pulmonary disease and interleukin-17A expression in nasal tissues of patients with chronic
overlap syndrome. Int J Chron Obstruct Pulmon Dis 2015;10:947- rhinosinusitis and asthma. Medicine (Baltimore) 2016;95:e5432.
54.pmid:26028967. doi:10.1097/MD.0000000000005432 pmid:27893686.
68 Lange P, Çolak Y, Ingebrigtsen TS, Vestbo J, Marott JL. Long-term prognosis 90 Ravensberg AJ, Slats AM, van Wetering S, et al. CD8(+) T cells
of asthma, chronic obstructive pulmonary disease, and asthma- characterize early smoking-related airway pathology in patients
chronic obstructive pulmonary disease overlap in the Copenhagen with asthma. Respir Med 2013;107:959-66. doi:10.1016/j.
City Heart study: a prospective population-based analysis. Lancet rmed.2013.03.018 pmid:23639272.
Respir Med 2016;4:454-62. doi:10.1016/S2213-2600(16)00098- 91 Saetta M, Baraldo S, Corbino L, et al. CD8+ve cells in the lungs
9 pmid:27061878. of smokers with chronic obstructive pulmonary disease.
69 Wurst KE, St Laurent S, Hinds D, Davis KJ. Disease Burden of Am J Respir Crit Care Med 1999;160:711-7. doi:10.1164/
Patients with Asthma/COPD Overlap in a US Claims Database: ajrccm.160.2.9812020 pmid:10430750.
Impact of ICD-9 Coding-based Definitions. COPD 2017;14:200-9. 92 O’Shaughnessy TC, Ansari TW, Barnes NC, Jeffery PK. Inflammation
doi:10.1080/15412555.2016.1257598 pmid:28103123. in bronchial biopsies of subjects with chronic bronchitis: inverse
70 Gerhardsson de Verdier M, Andersson M, Kern DM, Zhou S, Tunceli O. relationship of CD8+ T lymphocytes with FEV1. Am J Respir Crit Care Med
Asthma and Chronic Obstructive Pulmonary Disease Overlap Syndrome: 1997;155:852-7. doi:10.1164/ajrccm.155.3.9117016 pmid:9117016.
Doubled Costs Compared with Patients with Asthma Alone. Value Health 93 den Otter I, Willems LN, van Schadewijk A, et al. Lung function decline
2015;18:759-66. doi:10.1016/j.jval.2015.04.010 pmid:26409602. in asthma patients with elevated bronchial CD8, CD4 and CD3 cells.
71 Rhee CK, Yoon HK, Yoo KH, et al. Medical utilization and cost Eur Respir J 2016;48:393-402. doi:10.1183/13993003.01525-
in patients with overlap syndrome of chronic obstructive 2015 pmid:27230446.
pulmonary disease and asthma. COPD 2014;11:163-70. 94 Boulet LP, Turcotte H, Turcot O, Chakir J. Airway inflammation in asthma with
doi:10.3109/15412555.2013.831061 pmid:24111662. incomplete reversibility of airflow obstruction. Respir Med 2003;97:739-
72 Sadatsafavi M, Tavakoli H, Kendzerska T, et al. Canadian Respiratory 44. doi:10.1053/rmed.2003.1491 pmid:12814164.
Research Network. History of Asthma in Patients with Chronic Obstructive 95 Moerloose KB, Pauwels RA, Joos GF. Short-term cigarette smoke
Pulmonary Disease. A Comparative Study of Economic Burden. Ann Am exposure enhances allergic airway inflammation in mice. Am J Respir
Thorac Soc 2016;13:188-96.pmid:26599154. Crit Care Med 2005;172:168-72. doi:10.1164/rccm.200409-
73 Yamauchi Y, Yasunaga H, Matsui H, et al. Comparison of in-hospital 1174OC pmid:15831841.
mortality in patients with COPD, asthma and asthma-COPD overlap 96 Botelho FM, Llop-Guevara A, Trimble NJ, et al. Cigarette smoke differentially
exacerbations. Respirology 2015;20:940-6. doi:10.1111/ affects eosinophilia and remodeling in a model of house dust mite asthma.
resp.12556 pmid:25998444. Am J Respir Cell Mol Biol 2011;45:753-60. doi:10.1165/rcmb.2010-
74 Sluiter HJ, Koëter GH, de Monchy JG, Postma DS, de Vries K, Orie NG. The 0404OC pmid:21317378.
Dutch hypothesis (chronic non-specific lung disease) revisited. Eur Respir 97 Kumar S, Lanckacker E, Dentener M, et al. Aggravation of Allergic
J 1991;4:479-89.pmid:1855577. Airway Inflammation by Cigarette Smoke in Mice Is CD44-
75  American Thoracic Society. Standards for the diagnosis and care of Dependent. PLoS One 2016;11:e0151113. doi:10.1371/journal.
patients with chronic obstructive pulmonary disease. Am J Respir Crit Care pone.0151113 pmid:26999446.
Med 1995;152:S77-121.pmid:7582322. 98 Tilp C, Bucher H, Haas H, Duechs MJ, Wex E, Erb KJ. Effects of conventional
76 O’Donnell DE, Laveneziana P. Physiology and consequences tobacco smoke and nicotine-free cigarette smoke on airway inflammation,
of lung hyperinflation in COPD. Eur Respir Rev 2006;15:61- airway remodelling and lung function in a triple allergen model of
7doi:10.1183/09059180.00010002. severe asthma. Clin Exp Allergy 2016;46:957-72. doi:10.1111/
77 Gelb AF, Yamamoto A, Verbeken EK, Nadel JA. Unraveling the cea.12665 pmid:26502779.
Pathophysiology of the Asthma-COPD Overlap Syndrome: Unsuspected 99 Lefaudeux D, De Meulder B, Loza MJ, et al. U-BIOPRED Study Group.
Mild Centrilobular Emphysema Is Responsible for Loss of Lung Elastic U-BIOPRED clinical adult asthma clusters linked to a subset of sputum
Recoil in Never Smokers With Asthma With Persistent Expiratory omics. J Allergy Clin Immunol 2017;139:1797-807. doi:10.1016/j.
Airflow Limitation. Chest 2015;148:313-20. doi:10.1378/chest.14- jaci.2016.08.048 pmid:27773852.
2483 pmid:25950858. 100 Rice MB, Ljungman PL, Wilker EH, et al. Long-term exposure to traffic
78 Mauad T, Silva LF, Santos MA, et al. Abnormal alveolar attachments emissions and fine particulate matter and lung function decline in the
with decreased elastic fiber content in distal lung in fatal asthma. Am J Framingham heart study. Am J Respir Crit Care Med 2015;191:656-64.
Respir Crit Care Med 2004;170:857-62. doi:10.1164/rccm.200403- doi:10.1164/rccm.201410-1875OC pmid:25590631.
305OC pmid:15151920. 101 Adam M, Schikowski T, Carsin AE, et al. Adult lung function
79 Thomson NC, Chaudhuri R, Livingston E. Asthma and and long-term air pollution exposure. ESCAPE: a multicentre
cigarette smoking. Eur Respir J 2004;24:822-33. cohort study and meta-analysis. Eur Respir J 2015;45:38-50.
doi:10.1183/09031936.04.00039004 pmid:15516679. doi:10.1183/09031936.00130014 pmid:25193994.
80 Yii AC, Tan GL, Tan KL, Lapperre TS, Koh MS. Fixed airways obstruction 102 Gauderman WJ, Urman R, Avol E, et al. Association of improved air quality
among patients with severe asthma: findings from the Singapore General with lung development in children. N Engl J Med 2015;372:905-13.
Hospital-Severe Asthma Phenotype Study. BMC Pulm Med 2014;14:191. doi:10.1056/NEJMoa1414123 pmid:25738666.
doi:10.1186/1471-2466-14-191 pmid:25467558. 103 Zheng XY, Ding H, Jiang LN, et al. Association between Air
81 Tan DJ, Walters EH, Perret JL, et al. Clinical and functional differences Pollutants and Asthma Emergency Room Visits and Hospital
between early-onset and late-onset adult asthma: a population- Admissions in Time Series Studies: A Systematic Review and Meta-
based Tasmanian Longitudinal Health Study. Thorax 2016;71:981-7. Analysis. PLoS One 2015;10:e0138146. doi:10.1371/journal.
doi:10.1136/thoraxjnl-2015-208183 pmid:27301974. pone.0138146 pmid:26382947.

For personal use only 12 of 14


STAT E O F T H E A RT R E V I E W

104 Altzibar JM, Tamayo-Uria I, De Castro V, et al. Epidemiology of asthma 129 Hogg JC, Chu F, Utokaparch S, et al. The nature of small-airway obstruction
exacerbations and their relation with environmental factors in the in chronic obstructive pulmonary disease. N Engl J Med 2004;350:2645-
Basque Country. Clin Exp Allergy 2015;45:1099-108. doi:10.1111/ 53. doi:10.1056/NEJMoa032158 pmid:15215480.
cea.12419 pmid:25258133. 130 Kuwano K, Bosken CH, Paré PD, Bai TR, Wiggs BR, Hogg JC. Small
105 Esposito S, Galeone C, Lelii M, et al. Impact of air pollution on respiratory airways dimensions in asthma and in chronic obstructive pulmonary
diseases in children with recurrent wheezing or asthma. BMC Pulm Med disease. Am Rev Respir Dis 1993;148:1220-5. doi:10.1164/
2014;14:130. doi:10.1186/1471-2466-14-130 pmid:25098250. ajrccm/148.5.1220 pmid:8239157.
106 Perez L, Declercq C, Iñiguez C, et al. Chronic burden of 131 McDonough JE, Yuan R, Suzuki M, et al. Small-airway obstruction and
near-roadway traffic pollution in 10 European cities emphysema in chronic obstructive pulmonary disease. N Engl J Med
(APHEKOM network). Eur Respir J 2013;42:594-605. 2011;365:1567-75. doi:10.1056/NEJMoa1106955 pmid:22029978.
doi:10.1183/09031936.00031112 pmid:23520318. 132 Postma DS, Kerstjens HA. Characteristics of airway hyperresponsiveness
107 Weichenthal SA, Lavigne E, Evans GJ, Godri Pollitt KJ, Burnett RT. in asthma and chronic obstructive pulmonary disease. Am J Respir
Fine Particulate Matter and Emergency Room Visits for Respiratory Crit Care Med 1998;158:S187-92. doi:10.1164/ajrccm.158.
Illness. Effect Modification by Oxidative Potential. Am J Respir supplement_2.13tac170 pmid:9817744.
Crit Care Med 2016;194:577-86. doi:10.1164/rccm.201512- 133 Jones RL, Noble PB, Elliot JG, James AL. Airway remodelling in COPD:
2434OC pmid:26963193. It’s not asthma!Respirology 2016;21:1347-56. doi:10.1111/
108 Nadeau K, McDonald-Hyman C, Noth EM, et al. Ambient air pollution resp.12841 pmid:27381663.
impairs regulatory T-cell function in asthma. J Allergy Clin Immunol 134 Stănescu D, Sanna A, Veriter C, et al. Airways obstruction, chronic
2010;126:845-52. doi:10.1016/j.jaci.2010.08.008 pmid:20920773. expectoration, and rapid decline of FEV1 in smokers are associated
109 Hollingsworth JW, Maruoka S, Li Z, et al. Ambient ozone primes pulmonary with increased levels of sputum neutrophils. Thorax 1996;51:267-71.
innate immunity in mice. J Immunol 2007;179:4367-75. doi:10.4049/ doi:10.1136/thx.51.3.267 pmid:8779129.
jimmunol.179.7.4367 pmid:17878331. 135 Hodge G, Nairn J, Holmes M, Reynolds PN, Hodge S. Increased
110 Bowatte G, Lodge CJ, Lowe AJ, et al. Do Variants in GSTs Modify the intracellular T helper 1 proinflammatory cytokine production in peripheral
Association between Traffic Air Pollution and Asthma in Adolescence?Int J blood, bronchoalveolar lavage and intraepithelial T cells of COPD
Mol Sci 2016;17:485. doi:10.3390/ijms17040485 pmid:27043549. subjects. Clin Exp Immunol 2007;150:22-9. doi:10.1111/j.1365-
111 Bates JT, Weber RJ, Abrams J, et al. Reactive Oxygen Species 2249.2007.03451.x pmid:17614970.
Generation Linked to Sources of Atmospheric Particulate Matter and 136 Singh D, Kolsum U, Brightling CE, Locantore N, Agusti A, Tal-
Cardiorespiratory Effects. Environ Sci Technol 2015;49:13605-12. Singer R. ECLIPSE investigators. Eosinophilic inflammation in COPD:
doi:10.1021/acs.est.5b02967 pmid:26457347. prevalence and clinical characteristics. Eur Respir J 2014;44:1697-700.
112 Castro-Giner F, Künzli N, Jacquemin B, et al. Traffic-related air pollution, doi:10.1183/09031936.00162414 pmid:25323230.
oxidative stress genes, and asthma (ECHRS). Environ Health Perspect 137 Christenson SA, Steiling K, van den Berge M, et al. Asthma-COPD overlap.
2009;117:1919-24. doi:10.1289/ehp.0900589 pmid:20049212. Clinical relevance of genomic signatures of type 2 inflammation in chronic
113 Liu L, Poon R, Chen L, et al. Acute effects of air pollution on pulmonary obstructive pulmonary disease. Am J Respir Crit Care Med 2015;191:758-
function, airway inflammation, and oxidative stress in asthmatic 66. doi:10.1164/rccm.201408-1458OC pmid:25611785.
children. Environ Health Perspect 2009;117:668-74. doi:10.1289/ 138 Ghebre MA, Bafadhel M, Desai D, et al. Biological clustering supports
ehp.11813 pmid:19440509. both “Dutch” and “British” hypotheses of asthma and chronic
114 Klümper C, Krämer U, Lehmann I, et al. GINIplus and LISAplus study obstructive pulmonary disease. J Allergy Clin Immunol 2015;135:63-72.
groups. Air pollution and cytokine responsiveness in asthmatic and doi:10.1016/j.jaci.2014.06.035 pmid:25129678.
non-asthmatic children. Environ Res 2015;138:381-90. doi:10.1016/j. 139 Postma DS, Weiss ST, van den Berge M, Kerstjens HA, Koppelman GH.
envres.2015.02.034 pmid:25769127. Revisiting the Dutch hypothesis. J Allergy Clin Immunol 2015;136:521-9.
115 Brandt EB, Kovacic MB, Lee GB, et al. Diesel exhaust particle doi:10.1016/j.jaci.2015.06.018 pmid:26343936.
induction of IL-17A contributes to severe asthma. J Allergy 140 Kong X, Cho MH, Anderson W, et al. ECLIPSE Study NETT Investigators.
Clin Immunol 2013;132:1194-204. doi:10.1016/j. Genome-wide association study identifies BICD1 as a susceptibility gene
jaci.2013.06.048 pmid:24060272. for emphysema. Am J Respir Crit Care Med 2011;183:43-9. doi:10.1164/
116 Salam MT, Byun HM, Lurmann F, et al. Genetic and epigenetic variations in rccm.201004-0541OC pmid:20709820.
inducible nitric oxide synthase promoter, particulate pollution, and exhaled 141 Kottyan LC, Collier AR, Cao KH, et al. Eosinophil viability is
nitric oxide levels in children. J Allergy Clin Immunol 2012;129:232-9.e1, increased by acidic pH in a cAMP- and GPR65-dependent manner.
7. doi:10.1016/j.jaci.2011.09.037 pmid:22055874. Blood 2009;114:2774-82. doi:10.1182/blood-2009-05-
117 Somineni HK, Zhang X, Biagini Myers JM, et al. Ten-eleven translocation 1 220681 pmid:19641187.
(TET1) methylation is associated with childhood asthma and traffic-related 142 Barnes PJ. Therapeutic approaches to asthma-chronic obstructive
air pollution. J Allergy Clin Immunol 2016;137:797-805. doi:10.1016/j. pulmonary disease overlap syndromes. J Allergy Clin Immunol
jaci.2015.10.021 pmid:26684294. 2015;136:531-45. doi:10.1016/j.jaci.2015.05.052 pmid:26343937.
118 Gold WM, Kaufman HS, Nadel JA. Elastic recoil of the lungs in chronic 143 Miravitlles M, Soler-Cataluña JJ, Calle M, et al. Spanish Society of
asthmatic patients before and after therapy. J Appl Physiol 1967;23:433- Pulmonology and Thoracic Surgery. Spanish COPD Guidelines (GesEPOC):
8.pmid:6053669. pharmacological treatment of stable COPD. Arch Bronconeumol
119 Woolcock AJ, Read J. The static elastic properties of the lungs in asthma. 2012;48:247-57.pmid:22561012.
Am Rev Respir Dis 1968;98:788-94.pmid:5683472. 144 O’Donnell DE, Aaron S, Bourbeau J, et al. Canadian Thoracic Society
120 Gelb AF, Licuanan J, Shinar CM, Zamel N. Unsuspected loss of lung recommendations for management of chronic obstructive pulmonary
elastic recoil in chronic persistent asthma. Chest 2002;121:715-21. disease - 2007 update. Can Respir J 2007;14(Suppl B):5-32B.
doi:10.1378/chest.121.3.715 pmid:11888951. doi:10.1155/2007/830570 pmid:17885691.
121 McCarthy DS, Sigurdson M. Lung elastic recoil and reduced airflow in 145 Lim HS, Choi SM, Lee J, et al. Responsiveness to inhaled corticosteroid
clinically stable asthma. Thorax 1980;35:298-302. doi:10.1136/ treatment in patients with asthma-chronic obstructive pulmonary disease
thx.35.4.298 pmid:7434273. overlap syndrome. Ann Allergy Asthma Immunol 2014;113:652-7.
122 Gelb AF, Yamamoto A, Mauad T, Kollin J, Schein MJ, Nadel JA. Unsuspected doi:10.1016/j.anai.2014.08.021 pmid:25280465.
mild emphysema in nonsmoking patients with chronic asthma with 146 Feng JX, Lin Y, Lin J, et al. Relationship between Fractional Exhaled Nitric
persistent airway obstruction. J Allergy Clin Immunol 2014;133:263-5. Oxide Level and Efficacy of Inhaled Corticosteroid in Asthma-COPD Overlap
e1, 3. doi:10.1016/j.jaci.2013.09.045 pmid:24290280. Syndrome Patients with Different Disease Severity. J Korean Med Sci
123 Ingram JL, Slade D, Church TD, et al. Role of Matrix Metalloproteinases-1 2017;32:439-47. doi:10.3346/jkms.2017.32.3.439 pmid:28145647.
and -2 in Interleukin-13-Suppressed Elastin in Airway Fibroblasts 147 Lee SY, Park HY, Kim EK, et al. Combination therapy of inhaled steroids
in Asthma. Am J Respir Cell Mol Biol 2016;54:41-50. doi:10.1165/ and long-acting beta2-agonists in asthma-COPD overlap syndrome. Int
rcmb.2014-0290OC pmid:26074138. J Chron Obstruct Pulmon Dis 2016;11:2797-803. doi:10.2147/COPD.
124 Itabashi S, Fukushima T, Aikawa T, et al. Allergic sensitization in elderly S114964 pmid:27877033.
patients with chronic obstructive pulmonary disease. Respiration 148 Nixon J, Newbold P, Mustelin T, Anderson GP, Kolbeck R.
1990;57:384-8. doi:10.1159/000195876 pmid:2099572. Monoclonal antibody therapy for the treatment of asthma
125 Jamieson DB, Matsui EC, Belli A, et al. Effects of allergic phenotype and chronic obstructive pulmonary disease with eosinophilic
on respiratory symptoms and exacerbations in patients with chronic inflammation. Pharmacol Ther 2017;169:57-77. doi:10.1016/j.
obstructive pulmonary disease. Am J Respir Crit Care Med 2013;188:187- pharmthera.2016.10.016 pmid:27773786.
92. doi:10.1164/rccm.201211-2103OC pmid:23668455. 149 Maltby S, Gibson PG, Powell H, McDonald VM. Omalizumab
126 Gottlieb DJ, Sparrow D, O’Connor GT, Weiss ST. Skin test reactivity to Treatment Response in a Population With Severe Allergic Asthma
common aeroallergens and decline of lung function. The Normative and Overlapping COPD. Chest 2017;151:78-89. doi:10.1016/j.
Aging Study. Am J Respir Crit Care Med 1996;153:561-6. doi:10.1164/ chest.2016.09.035 pmid:27742181.
ajrccm.153.2.8564098 pmid:8564098. 150 Tat TS, Cilli A. Omalizumab treatment in asthma-COPD
127 Cockcroft DW, Wenzel S. Airway hyperresponsiveness and chronic overlap syndrome. J Asthma 2016;53:1048-50.
obstructive pulmonary disease outcomes. J Allergy Clin Immunol doi:10.1080/02770903.2016.1178281 pmid:27144514.
2016;138:1580-1. doi:10.1016/j.jaci.2016.06.039 pmid:27516214. 151 Yalcin AD, Celik B, Yalcin AN. Omalizumab (anti-IgE) therapy in the
128 Ramsdell JW, Nachtwey FJ, Moser KM. Bronchial hyperreactivity in asthma-COPD overlap syndrome (ACOS) and its effects on circulating
chronic obstructive bronchitis. Am Rev Respir Dis 1982;126:829- cytokine levels. Immunopharmacol Immunotoxicol 2016;38:253-6.
32.pmid:7149448. doi:10.3109/08923973.2016.1173057 pmid:27121601.

For personal use only 13 of 14


STAT E O F T H E A RT R E V I E W

152 Varricchi G, Bagnasco D, Borriello F, Heffler E, Canonica GW. 169 Sutherland ER, King TS, Icitovic N, et al. National Heart, Lung and Blood


Interleukin-5 pathway inhibition in the treatment of eosinophilic Institute’s Asthma Clinical Research Network. A trial of clarithromycin for
respiratory disorders: evidence and unmet needs. Curr Opin the treatment of suboptimally controlled asthma. J Allergy Clin Immunol
Allergy Clin Immunol 2016;16:186-200. doi:10.1097/ 2010;126:747-53. doi:10.1016/j.jaci.2010.07.024 pmid:20920764.
ACI.0000000000000251 pmid:26859368. 170 Kostadima E, Tsiodras S, Alexopoulos EI, et al. Clarithromycin
153 Ortega HG, Liu MC, Pavord ID, et al. MENSA Investigators. Mepolizumab reduces the severity of bronchial hyperresponsiveness
treatment in patients with severe eosinophilic asthma. N Engl J Med in patients with asthma. Eur Respir J 2004;23:714-7.
2014;371:1198-207. doi:10.1056/NEJMoa1403290 pmid:25199059. doi:10.1183/09031936.04.00118404 pmid:15176685.
154 Castro M, Wenzel SE, Bleecker ER, et al. Benralizumab, an anti-interleukin 171 Black PN, Blasi F, Jenkins CR, et al. Trial of roxithromycin in subjects
5 receptor α monoclonal antibody, versus placebo for uncontrolled with asthma and serological evidence of infection with Chlamydia
eosinophilic asthma: a phase 2b randomised dose-ranging study. Lancet pneumoniae. Am J Respir Crit Care Med 2001;164:536-41.
Respir Med 2014;2:879-90. doi:10.1016/S2213-2600(14)70201- doi:10.1164/ajrccm.164.4.2011040 pmid:11520711.
2 pmid:25306557. 172 Amayasu H, Yoshida S, Ebana S, et al. Clarithromycin
155 Bel EH, Wenzel SE, Thompson PJ, et al. SIRIUS Investigators. Oral suppresses bronchial hyperresponsiveness associated with
glucocorticoid-sparing effect of mepolizumab in eosinophilic eosinophilic inflammation in patients with asthma. Ann Allergy
asthma. N Engl J Med 2014;371:1189-97. doi:10.1056/ Asthma Immunol 2000;84:594-8. doi:10.1016/S1081-
NEJMoa1403291 pmid:25199060. 1206(10)62409-X pmid:10875487.
156 Castro M, Zangrilli J, Wechsler ME, et al. Reslizumab for inadequately 173 Shoji T, Yoshida S, Sakamoto H, Hasegawa H, Nakagawa H, Amayasu H.
controlled asthma with elevated blood eosinophil counts: results from Anti-inflammatory effect of roxithromycin in patients with aspirin-
two multicentre, parallel, double-blind, randomised, placebo-controlled, intolerant asthma. Clin Exp Allergy 1999;29:950-6. doi:10.1046/
phase 3 trials. Lancet Respir Med 2015;3:355-66. doi:10.1016/S2213- j.1365-2222.1999.00551.x pmid:10383596.
2600(15)00042-9 pmid:25736990. 174 Strunk RC, Bacharier LB, Phillips BR, et al. CARE Network. Azithromycin
157 Brightling CE, Bleecker ER, Panettieri RA Jr, et al. Benralizumab for or montelukast as inhaled corticosteroid-sparing agents in moderate-to-
chronic obstructive pulmonary disease and sputum eosinophilia: a severe childhood asthma study. J Allergy Clin Immunol 2008;122:1138-
randomised, double-blind, placebo-controlled, phase 2a study. Lancet 44. doi:10.1016/j.jaci.2008.09.028 pmid:18951618.
Respir Med 2014;2:891-901. doi:10.1016/S2213-2600(14)70187- 175 Kew KM, Undela K, Kotortsi I, Ferrara G. Macrolides for chronic asthma.
0 pmid:25208464. Cochrane Database Syst Rev 2015;9:CD002997.pmid:26371536.
158 Wenzel S, Castro M, Corren J, et al. Dupilumab efficacy and safety in adults 176 Kerstjens HA, Engel M, Dahl R, et al. Tiotropium in asthma
with uncontrolled persistent asthma despite use of medium-to-high-dose poorly controlled with standard combination therapy.
inhaled corticosteroids plus a long-acting β2 agonist: a randomised N Engl J Med 2012;367:1198-207. doi:10.1056/
double-blind placebo-controlled pivotal phase 2b dose-ranging trial. NEJMoa1208606 pmid:22938706.
Lancet 2016;388:31-44. doi:10.1016/S0140-6736(16)30307- 177 Szefler SJ, Murphy K, Harper T 3rd, et al. A phase III randomized controlled
5 pmid:27130691. trial of tiotropium add-on therapy in children with severe symptomatic
159 Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent asthma asthma. J Allergy Clin Immunol 2017;pii:S0091-6749(17)30218-
with elevated eosinophil levels. N Engl J Med 2013;368:2455-66. X.pmid:28189771.
doi:10.1056/NEJMoa1304048 pmid:23688323. 178 Hamelmann E, Bernstein JA, Vandewalker M, et al. A randomised
160 Hanania NA, Noonan M, Corren J, et al. Lebrikizumab in moderate- controlled trial of tiotropium in adolescents with severe
to-severe asthma: pooled data from two randomised placebo- symptomatic asthma. Eur Respir J 2017;49:1601100.
controlled studies. Thorax 2015;70:748-56. doi:10.1136/ doi:10.1183/13993003.01100-2016 pmid:27811070.
thoraxjnl-2014-206719 pmid:26001563. 179 Kerstjens HA, Moroni-Zentgraf P, Tashkin DP, et al. Tiotropium improves
161 Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in lung function, exacerbation rate, and asthma control, independent of
adults with asthma. N Engl J Med 2011;365:1088-98. doi:10.1056/ baseline characteristics including age, degree of airway obstruction,
NEJMoa1106469 pmid:21812663. and allergic status. Respir Med 2016;117:198-206. doi:10.1016/j.
162 Brightling CE, Chanez P, Leigh R, et al. Efficacy and safety of tralokinumab rmed.2016.06.013 pmid:27492532.
in patients with severe uncontrolled asthma: a randomised, double-blind, 180 Timmer W, Moroni-Zentgraf P, Cornelissen P, Unseld A, Pizzichini E, Buhl R.
placebo-controlled, phase 2b trial. Lancet Respir Med 2015;3:692-701. Once-daily tiotropium Respimat(®) 5 μg is an efficacious
doi:10.1016/S2213-2600(15)00197-6 pmid:26231288. 24-h bronchodilator in adults with symptomatic
163 Izuhara K, Ohta S, Ono J. Using Periostin as a Biomarker in the Treatment asthma. Respir Med 2015;109:329-38. doi:10.1016/j.
of Asthma. Allergy Asthma Immunol Res 2016;8:491-8. doi:10.4168/ rmed.2014.12.005 pmid:25661281.
aair.2016.8.6.491 pmid:27582399. 181 Kerstjens HA, Casale TB, Bleecker ER, et al. Tiotropium or salmeterol as
164 Culić O, Eraković V, Cepelak I, et al. Azithromycin modulates neutrophil add-on therapy to inhaled corticosteroids for patients with moderate
function and circulating inflammatory mediators in healthy human symptomatic asthma: two replicate, double-blind, placebo-controlled,
subjects. Eur J Pharmacol 2002;450:277-89. doi:10.1016/S0014- parallel-group, active-comparator, randomised trials. Lancet Respir
2999(02)02042-3 pmid:12208321. Med 2015;3:367-76. doi:10.1016/S2213-2600(15)00031-
165 Albert RK, Connett J, Bailey WC, et al. COPD Clinical Research Network. 4 pmid:25682232.
Azithromycin for prevention of exacerbations of COPD. N Engl J Med 182 Peters SP, Kunselman SJ, Icitovic N, et al. National Heart, Lung, and
2011;365:689-98. doi:10.1056/NEJMoa1104623 pmid:21864166. Blood Institute Asthma Clinical Research Network. Tiotropium bromide
166 Brusselle GG, Vanderstichele C, Jordens P, et al. Azithromycin for step-up therapy for adults with uncontrolled asthma. N Engl J Med
prevention of exacerbations in severe asthma (AZISAST): a multicentre 2010;363:1715-26. doi:10.1056/NEJMoa1008770 pmid:20979471.
randomised double-blind placebo-controlled trial. Thorax 2013;68:322- 183 Price D, Kaplan A, Jones R, et al. Long-acting muscarinic antagonist
9. doi:10.1136/thoraxjnl-2012-202698 pmid:23291349. use in adults with asthma: real-life prescribing and outcomes of
167 Stokholm J, Chawes BL, Vissing NH, et al. Azithromycin for episodes with add-on therapy with tiotropium bromide. J Asthma Allergy 2015;8:1-
asthma-like symptoms in young children aged 1-3 years: a randomised, 13.pmid:25609985.
double-blind, placebo-controlled trial. Lancet Respir Med 2016;4:19-26. 184 GlaxoSmithKline. GSK announces headline phase III results of
doi:10.1016/S2213-2600(15)00500-7 pmid:26704020. mepolizumab in patients with severe chronic obstructive pulmonary
168 Cameron EJ, Chaudhuri R, Mair F, et al. Randomised controlled trial of disease. 2017. https://www.gsk.com/en-gb/media/press-releases/
azithromycin in smokers with asthma. Eur Respir J 2013;42:1412-5. gsk-announces-headline-phase-iii-results-of-mepolizumab-in-patients-
doi:10.1183/09031936.00093913 pmid:24036246. with-severe-chronic-obstructive-pulmonary-disease/.

For personal use only 14 of 14

Das könnte Ihnen auch gefallen