Sie sind auf Seite 1von 18

609251

research-article2015
TAJ0010.1177/2040622315609251Therapeutic Advances in Chronic DiseaseL George and CE Brightling

Therapeutic Advances in Chronic Disease Review

Eosinophilic airway inflammation:


Ther Adv Chronic Dis

2016, Vol. 7(1) 3451

role in asthma and chronic obstructive DOI: 10.1177/


2040622315609251

pulmonary disease
The Author(s), 2015.
Reprints and permissions:
http://www.sagepub.co.uk/
journalsPermissions.nav

Leena George and Christopher E. Brightling

Abstract: The chronic lung diseases, asthma and chronic obstructive pulmonary disease
(COPD), are common affecting over 500 million people worldwide and causing substantial
morbidity and mortality. Asthma is typically associated with Th2-mediated eosinophilic
airway inflammation, in contrast to neutrophilic inflammation observed commonly in COPD.
However, there is increasing evidence that the eosinophil might play an important role in
1040% of patients with COPD. Consistently in both asthma and COPD a sputum eosinophilia
is associated with a good response to corticosteroid therapy and tailored strategies aimed to
normalize sputum eosinophils reduce exacerbation frequency and severity. Advances in our
understanding of the multistep paradigm of eosinophil recruitment to the airway, and the
consequence of eosinophilic inflammation, has led to the development of new therapies to
target these molecular pathways. In this article we discuss the mechanisms of eosinophilic
trafficking, the tools to assess eosinophilic airway inflammation in asthma and COPD during
stable disease and exacerbations and review current and novel anti-eosinophilic treatments.

Keywords: ACOS, anti-CRTh2, anti-IL5, anti-IL-13, anti-IL4R, asthma, biomarker, COPD,


corticosteroid, eosinophil

Introduction (Th2)-mediated immune response is the hallmark Correspondence to:


Christopher E. Brightling,
Asthma and chronic obstructive pulmonary dis- of airway inflammation in asthma, but importantly FRCP PhD
ease (COPD) cause considerable morbidity eosinophilic inflammation can occur independent Institute for Lung Health,
Clinical Science Wing,
affecting over 500 million people worldwide and of allergy and the presence of eosinophilic inflam- University Hospital of
managing these conditions consumes substantial mation is neither necessary nor sufficient for the Leicester, Leicester LE3
9QP, UK
healthcare resources exceeding 56 billion per development of asthma [Eltboli and Brightling, ceb17@le.ac.uk
year in the European Union [Burrowes et al. 2013]. Conversely, COPD is typically associated Leena George, MRCP
2014]. Both conditions are characterized by air- with T helper 1 lymphocytes (Th1)-mediated Institute for Lung Health,
NIHR Respiratory
flow obstruction, which is typically variable and immunity with a neutrophilic response [Stanescu Biomedical Research
reversible in asthma, but fixed in COPD. Similarly etal. 1996] often in association with bacterial col- Unit, Department of
Infection, Immunity and
the symptoms in asthma are usually intermittent, onization. However, this probably represents only Inflammation, University
although persistent in more severe disease, half of COPD patients and in a subgroup of of Leicester and University
Hospitals of Leicester NHS
whereas in COPD symptoms occur daily with 1040% eosinophilic airway inflammation is a fea- Trust, Leicester, UK
reduced exercise capacity and respiratory failure ture [Brightling et al. 2000, 2005; Eltboli et al.
occurring as the disease progresses. Both condi- 2014; Leigh et al. 2006; Pizzichini et al. 1998;
tions are characterized by exacerbations whereby Saetta etal. 1994]. Therefore, eosinophilic inflam-
sufferers have worsening of their symptoms often mation cannot be assumed to be present or absent
precipitated by infection and these exacerbations dependent on the airway disease, but needs to
amplify the morbidity and contribute substan- be measured as part of the assessment and pheno-
tially to the mortality of these airway diseases. typing of airways disease. This is critical when
considering the potential immunopathogenic
Eosinophilic inflammation orchestrated by role of eosinophilic inflammation in disease; its
allergic sensitization and T helper 2 lymphocytes role as a biomarker to direct current therapies and

34 http://taj.sagepub.com
L George and CE Brightling

for the future development of eosinophil-directed 1996]. IL-4 and IL-13 have been found to increase
therapies. VCAM-1 and P-selectin expression [Patel, 1998;
Woltmann etal. 2000]. Recruitment to the airway
Herein, we shall discuss the mechanisms of eosin- is under the control of the chemokines CCL5, 7,
ophilic trafficking, the tools to assess eosinophil 11, 13, 15, 24 and 26 and their cognate receptor
airway inflammation, cross-sectional and longitu- CCR3 [Smit and Lukacs, 2006; Ying etal. 1997],
dinal analysis of eosinophilic airway inflammation which play a critical role together with chemo-
in asthma and COPD during stable disease and attractant receptor homologous molecule
exacerbations and review anti-eosinophilic treat- expressed on Th2 cells (CRTh2) and its ligand
ments including therapies in development. prostaglandin D2 (PGD2) [Hirai et al. 2001;
Mutalithas et al. 2010]. PGD2 acts both as an
eosinophil chemoattractant in its own right, but
Eosinophil biology: mechanisms of also augments the effects of the CCR3 chemokines
eosinophilic airway inflammation [Mesquita-Santos et al. 2006]. Activated mast
Eosinophils are granulocytic leukocytes first dis- cells are the predominant source of PGD2 with
covered by Heinrich Caro in 1874 and described release via IgE-dependent and independent acti-
by Paul Ehrlich in 1879 [Kay, 2014]. They con- vation [Lewis etal. 1982].
stitute 14% of circulating white cells and are dis-
tinguished phenotypically by their bilobed nuclei Eosinophils have four specific basic proteins that
and large acidophilic cytoplasmic granules. The include major basic protein (MBP), eosinophil
pathological role of eosinophils primarily occurs cationic protein (ECP), eosinophil peroxidase
in tissues and therefore a major focus has been to (EPO) and eosinophil derived neurotoxin (EDN),
outline the molecular mechanisms involved in which are stored in secondary granules. ECP has
selective eosinophil recruitment summarized in been found to attach to cell membrane altering
Figure 1 [Rosenberg etal. 2007; Wardlaw, 1999]. permeability as well as increasing production of
reactive oxygen species [Navarro etal. 2008]. MBP
Eosinophils are derived from pluripotent CD34+ was also found to cause alveolar epithelium cell
progenitor stem cells found in normal bone mar- lysis allowing for penetration of inhaled antigen
row. This differentiation occurs under the influ- [Ayars etal. 1985]. In combination these granules
ence of granulocyte-monocyte colony stimulating are cytotoxic and disrupt the protective pulmonary
factor (GM-CSF) and interleukin (IL)-3 in early epithelial barrier allowing further inflammatory
phases and IL-5 in the latter phases of differentia- responses to occur [Frigas etal. 1991]. Eosinophils
tion [Denburg, 1998]. Mature eosinophils are have also been characterized by the ability to store
released from bone marrow into circulation and preformed mediators as well as synthesize and
this is primarily regulated by IL-5. Eosinophils are secrete a number of pro-inflammatory cytokines,
transformed from a quiescent state to a state of chemokines and growth factors including IL-2,
increased hyper responsiveness by priming agents IL-3, IL-4, IL-5, IL-10, IL-12, IL-13, IL-16,
such as these cytokines IL-3, IL-5, GM-CSF IL-25, tumour necrosis factor, transforming
[Luijk etal. 2005]. The priming process does not growth factor (TGF) /, CCL5, CCL11, 13
fully activate the cell but increases its responsive- facilitating their key role as immunoregulators
ness to chemotaxis, degranulation and cytokine [Davoine and Lacy, 2014; Moqbel etal. 1994].
production [Fulkerson and Rothenberg, 2013].
As eosinophils flow in the blood stream they roll Thus, although eosinophils have an immunomod-
onto the bronchial vascular endothelium. ulatory role they are recruited into the airway
Infiltration of eosinophils into the airway will not orchestrated by other cells and can be considered
occur unless there is adhesion and transmigration largely a final important effector cell.
across the bronchial vascular endothelium. This
preferential adhesion of eosinophils from the
blood stream into various tissues results from spe- Phenotyping the heterogeneity of eosinophilic
cific interactions between integrins on surface of airway inflammation
eosinophils and adhesion receptors on the surface
of vascular endothelium, which include P-selectin/ How can you measure eosinophilic
P-selectin glycoprotein ligand-1 and very late acti- inflammation?
vation antigen/vascular cell adhesion molecule, Eosinophilic airway inflammation can be measured
VCAM-1 ligand [Fukuda etal. 1996; Symon etal. in the airway noninvasively by sputum analysis

http://taj.sagepub.com 35
Therapeutic Advances in Chronic Disease 7(1)

Figure 1. Eosinophil trafficking from the bone marrow to the airway.

and invasively by bronchoscopic sampling. interval (CI) 0.780.93] [Bafadhel et al. 2011;
Bronchoscopy enables investigators to sample Wagener etal. 2015] to detect a sputum eosino-
airway inflammation directly in the airway wall philia 3%. Blood eosinophils can be assessed in
by bronchial biopsy, in the large airway lumen most hospitals worldwide and thus presents a bio-
by proximal bronchial wash and in the smaller marker that has advantages due to its simplicity
distal airways with bronchoalveolar lavage. and availability. The relationship between blood
Bronchoscopic sampling is invasive costly, time and sputum eosinophils is complicated in obesity.
consuming and requires the need of experienced Recent evidence demonstrates that in obese asth-
investigators. The relationship between sputum matics the relationship is distinct with strong
and bronchoscopic sampling has been investi- associations between blood and tissue eosino-
gated in several studies and although they gener- philia, but a consistently reduced sputum eosino-
ally report correlations between compartments philia suggesting altered eosinophil trafficking in
these correlations are weak [Keatings etal. 1997; obesity [Desai etal. 2013].
Maestrelli etal. 1995; Rutgers etal. 2000]. The
use of sputum cytology to explore airway inflam- Interestingly, sputum cytology also reveals infor-
mation has been in existence since the first mation about longer term eosinophil exposure
descriptions by Ernst Leyden and Jean Charcot of beyond the simple sputum eosinophil differential
Charcot-Leyden crystals [Sakula, 1986] and was cell count. The apoptosis and subsequent removal
used clinically in the 1950s by Dr Morrow-Brown of dead cells by phagocytes (efferocytosis) is a
[Brown, 1958] and then optimized in the 1990s critical mechanism for the noninflammatory
by Freddy Hargreaves and colleagues [Pavord clearance of granulocytes including eosinophils.
et al. 1997]. Normal sputum eosinophil count Macrophage red hue due to eosin staining within
ranges in large populations have been defined. sputum macrophages reflects eosinophil efferocy-
A blood eosinophil count has been used as a sur- tosis and can be used as a biomarker of eosinophil
rogate for eosinophilic airway inflammation and load and its clearance [Eltboli etal. 2014; Kulkarni
to determine the intensity of eosinophilic inflam- etal. 2010].
mation in the blood compartment [Bafadhel etal.
2012; Wagener etal. 2015]. Studies have shown a Other biomarkers that are associated with eosino-
moderate correlation between blood eosinophil philic airway inflammation include fractional
count and sputum eosinophil count (r=0.6; exhaled nitric oxide (FeNO) or Th2-activation
p<0.0001) [Schleich et al. 2013] (r=0.59; marker, e.g. serum periostin [Berry etal. 2005].
p<0.001) [Wagener et al. 2015]. Using the Breath analysis is an attractive alternative as a
(ROC) curves the diagnostic accuracy of a blood near-patient test. Nitric oxide is produced by var-
eosinophil count was 0.85 [95% confidence ious cells including airway epithelial cells and

36 http://taj.sagepub.com
L George and CE Brightling

inflammatory cells under the action of inducible How common is eosinophilic inflammation in
NO synthase enzyme which converts L-arginine asthma and COPD and is it a stable phenotype?
to L-citrulline. FeNO levels are correlated with In asthma up to 80% of corticosteroid-nave
peripheral blood eosinophils [Zietkowski et al. patients and 50% of corticosteroid treated
2006] and sputum eosinophils particularly in patients have a sputum eosinophilia [Douwes
mild-to-moderate disease whereas the relation- etal. 2002; Eltboli and Brightling, 2013]. Short-
ship is more complex in severe disease. FeNO in term repeatability of a sputum eosinophilia is very
severe asthma was poorly related to blood eosino- good [Boorsma etal. 2007; Pizzichini etal. 1996].
phils as highlighted in a national registry audit in However, there is emerging evidence that in some
refractory asthma, which showed that though patients their inflammatory profile is stable
blood eosinophils levels decreased, FeNO levels whereas in others it is highly variable. McGrath
paradoxically increased in response to oral corti- and colleagues in a population of 995 asthma
costeroids [Sweeney etal. 2012]. Novel breath- patients reported that persistent eosinophilic
omic approaches also have promise to identify inflammation was less frequent than subjects with
different inflammatory phenotypes. an intermittent eosinophilia, 22% versus 31%
[McGrath etal. 2012]. Interestingly Newby and
Periostin is a matricellular protein secreted baso- colleagues [Newby et al. 2014a] have demon-
laterally from epithelial cells in response to vari- strated that in severe asthma those subjects with
ous stimuli including IL-13 and its levels are intermittent eosinophilia had increased rate of
highly correlated with Th2 signature [Woodruff lung function decline compared with those with-
etal. 2007]. Two major studies have highlighted out eosinophilic inflammation or persistent eosin-
apparently conflicting views on the correlation of ophilic inflammation.
periostin with eosinophilia. An observational
study found periostin to be a superior predictor of Further insights into how common eosinophilic
eosinophil inflammation in the airway [Jia et al. inflammation is in asthma can be derived from
2012] while a more recent a cross-sectional study corticosteroid withdrawal studies as emergence of
[Wagener etal. 2015] showed a strong correlation eosinophilic inflammation following treatment
of sputum eosinophils with blood eosinophils, but withdrawal in apparently noneosinophilic disease
failed to demonstrate a relationship between is informative. In a 1-year follow-up study of asth-
serum periostin and eosinophils in sputum. The matic patients in whom treatment was stepped-
most likely explanation for these apparent differ- down in the absence of a sputum eosinophilia,
ences are that former study investigated the sensi- persistent noneosinophilia was observed in 12.5%
tivity and specificity of periostin to identify an of patients [Kulkarni et al. 2010]. Interestingly,
airway eosinophilia using a composite score rather the macrophage red hue predicts the emergence
than specifically comparing the relationship of eosinophilic inflammation in treatment with-
between blood periostin and a sputum eosinophil drawal with continued corticosteroid step-down
count as performed in the later study. This sug- only possible in those subjects without evidence
gests that blood periostin might provide addi- of a sputum eosinophilia nor elevated macrophage
tional value as a biomarker of Th2 inflammation, red hue [Kulkarni et al. 2010]. This suggests
but is not directly comparable with either blood that in severe asthma the absence of an under-
or sputum eosinophilia. Periostin may be more lying eosinophilic inflammation is probably
useful as a marker to assess specific IL-13 activity uncommon.
and responses to anti IL-13 therapy [Corren etal.
2011]. IL-13 induces epithelial-derived periostin In COPD, although neutrophilic inflammation is
directly and is associated with upregulated FENO commonly reported, a sputum eosinophilia is pre-
expression, whereas the relationship between air- sent in 1040% of patients with COPD [Brightling
way eosinophilic inflammation and IL-13 is more etal. 2000; Leigh etal. 2006; Pizzichini etal. 1998;
complex and indirect such as via upregulation of Saetta etal. 1994] Similarly to asthma the repeat-
epithelial-derived chemoattractants. These varia- ability of eosinophilic inflammation is very good,
tions in biomarker measurements in various stud- but again there are three groups with either persis-
ies further highlights the heterogeneity and tent, variable or absent eosinophilic inflammation.
complexity of airway inflammation and the pos- In the ECLIPSE (Evaluation of COPD
sible need of measuring a number of biomarkers Longitudinally to Identify Predictive Surrogate
in conjunction with one another to accurately try End points) cohort repeated blood eosinophil
and determine a phenotype. counts were available in 1483 subjects over 3 years

http://taj.sagepub.com 37
Therapeutic Advances in Chronic Disease 7(1)

[Singh etal. 2014]. Using a cutoff for blood eosin- [Green et al. 2002b; Jatakanon et al. 2000; Siva
ophils of 2%, 37% of subjects had persistently et al. 2007]. However, the relationship between
elevated eosinophils, 49% were intermittent and eosinophilic inflammation and airflow obstruc-
the remainder were noneosinophilic. The persis- tion and airway hyper-responsiveness is weak
tently eosinophilic group were older, predomi- exemplified by the presence of eosinophilic
nantly male with milder disease and fewer inflammation in eosinophilic bronchitis without
symptoms. Emphysema progression was greater asthma [Gibson etal. 1989, 1995]. Indeed com-
in the non-eosinophilic group [Singh etal. 2014] parisons with this disease group have revealed the
and in contrast to severe asthma there were no dif- important role of mast cell localisation to the air-
ferences in lung function decline between groups. way smooth muscle bundle in driving disordered
airway physiology [Brightling et al. 2002].
To further inform the understanding of the het- Eosinophils also contribute to airway remodelling
erogeneity of airway disease unbiased statistical [Flood-Page etal. 2003; Kay etal. 2004]. Airway
approaches such as cluster analysis have been remodelling is a consequence of increased extra-
applied to large clinical datasets [Amelink et al. cellular matrix deposition, increased subepithelial
2013; Haldar et al. 2008; Newby et al. 2014b; mesenchymal cells and most importantly
Wardlaw et al. 2005]. Interestingly, these have increased airway smooth muscle mass, which is
underscored the importance of eosinophilic air- the major determinant of airflow obstruction.
way inflammation in asthma and COPD and per- Eosinophil-derived TGF- activates fibroblast
haps most importantly in asthma-COPD overlap release of matrix proteins and although increased
syndrome (ACOS). For example, Ghebre and airway smooth muscle mass was eosinophil
colleagues combined data from asthma and dependent in a mouse model of asthma [Humbles
COPD and identified three clusters [Ghebre etal. et al. 2004] whether the eosinophil is necessary
2015]. Cluster 1 consisted of asthma subjects for this critical feature of remodelling in human
with increase IL-5, IL-13 and CCL26 mediators disease is questionable as evidenced by interven-
and eosinophil predominance. Cluster 2 con- tion studies described later. Whereas in contrast,
sisted of an overlap between asthma and COPD reticular basement membrane thickening is asso-
with neutrophil predominance with 11% of ciated with eosinophilic inflammation in asthma,
patients with asthma having sputum eosinophilia. COPD and nonasthmatic eosinophilic bronchitis
Cluster 3 consisted mainly of COPD patients [Brightling etal. 2003; Eltboli etal. 2015; Siddiqui
with a mixed granulocytic airway inflammation. etal. 2008].
The differences seen between neutrophilic COPD
in cluster 2 and eosinophilic COPD in cluster 3
included the presence of increased bacterial colo- Is eosinophilic inflammation a feature of
nization in the former and increased CCL13 in asthma and COPD exacerbations?
the latter possibly explaining the observed airway The definition of asthma exacerbations vary
inflammation differences seen between these according to literature source and asthma severity.
clusters. Importantly few studies have investi- In summary asthma exacerbations are defined as a
gated cluster stability and predictably due to the decrease in peak flow with worsening of symptoms
variability of eosinophilic inflammation in some requiring the use of reliever treatment and or sys-
patients as described above cluster membership temic corticosteroids [Reddel etal. 2009; see also
can change overtime. This further highlights the http://www.ginasthma.org]. Severe asthma exac-
importance of considering the dynamics of eosin- erbations require 3 or more days of high-dose oral
ophilic inflammation within an individual to corticosteroids. GOLD (see http://www.goldcopd.
appreciate the relationship of this inflammatory org) have defined a COPD exacerbation as an
phenotype with natural history of disease and acute event characterised by worsening of the
response to therapy. patients respiratory symptoms that is beyond nor-
mal day-to-day variations and leads to a change in
medication. The differences in exacerbation defi-
Is eosinophilic inflammation related to nitions for asthma and COPD poses limitations in
disordered airway physiology and remodelling? comparisons between the diseases. However, both
Eosinophil-derived granule proteins and pro- asthma and COPD exacerbations are commonly
inflammatory mediators promote persistent associated with identification of pathogens usually
inflammation, which has been associated with viral although also bacterial particularly in
increased exacerbations and lung function decline COPD. However, pathogens are not identified at

38 http://taj.sagepub.com
L George and CE Brightling

exacerbations in 2050% of cases [Bafadhel etal. time, longitudinally in stable disease, at an exacer-
2011; Sethi and Murphy, 2008] either due to bation and in response to therapy. For example, in
insensitivity of assays or as a consequence of a lack the study of benralizumab in COPD described
of a role of new infections in some exacerbations. below beneficial effects were observed in terms of
Therefore, airway inflammation at exacerbations lung function, health status and exacerbations at
might represent a consequence of an acute envi- different baseline blood and sputum eosinophil
ronmental insult on an intrinsically unstable state counts [Brightling etal. 2014]. Thus, the applica-
reflecting an emergent phenomenon in a complex tion of different cutoffs to direct therapies and
system [Brightling, 2013]. monitor disease are likely to vary depending on
the intervention of interest together with consid-
Eosinophilic airway inflammation is present in eration of the potential health economic benefits
exacerbations of asthma and COPD, particularly for these interventions at different cutoffs. For this
in those subjects with eosinophilic inflammation in reason although normal ranges are established a
stable state. A high baseline sputum eosinophil single cutoff to direct all therapy is unlikely.
count is a predictor of loss of control of asthma and
an increased number of subsequent exacerbations
[Jatakanon etal. 2000]. Treatment strategies aim- Therapies that modulate eosinophilic
ing to maintain sputum eosinophil counts <23% inflammation
is associated with a significant decrease in the The importance of eosinophilic inflammation in
number of exacerbations as compared with treat- asthma and COPD and its role in different states
ments focused on clinical history or spirometry of diseases is perhaps best informed by response
[Brightling etal. 2000; Green etal. 2002a; Jayaram to therapies. We describe here current and emerg-
et al. 2006]. In COPD sputum eosinophilia has ing therapies targeting eosinophilic inflammation
been seen in 1040% of patients at baseline and consider their overall clinical effusiveness;
[Brightling etal. 2000; Pizzichini etal. 1998; Singh their impact upon eosinophilic inflammation and
etal. 2014] with higher eosinophilic inflammation consequent new insights into the relationship
seen at exacerbation states compared to stable between changes in eosinophilic inflammation
states [Saetta etal. 1994]. In a study aimed at phe- and clinical outcomes.
notyping COPD exacerbations and identifying
biomarkers a sputum eosinophilia was identified in
28% of exacerbations [Bafadhel etal. 2011]. The Corticosteroids
combination of sputum eosinophil count and mac- Inhaled and systemic corticosteroids are the main-
rophage red hue was used to categorize COPD stay anti-inflammatory therapy for asthma and
subjects into four groups according to high and COPD in stable disease and at exacerbations as
low sputum eosinophils (cutoff 3%) and high reflected in their central position in international
and low percentage macrophage red hue area (cut- guidelines supported by extensive meta-analyses
off >6%) [Eltboli et al. 2014]. The group with and Cochrane reviews of multiple randomized
high eosinophil count, but low red hue had the controlled trials [Edwards et al. 2010; Loymans
greatest fall in lung function during an exacerba- etal. 2014; Yang etal. 2012]. The mechanism of
tion and also increased exacerbation frequency. In action of corticosteroids has been studied exten-
this group macrophage efferocytosis is defective sively, but remains incompletely understood. In
contributing to persistent eosinophilic inflamma- brief, corticosteroids diffuse across the eosinophil
tion, which in turn is likely to impact upon the cell membrane and enter the cell cytoplasm where
severity of the exacerbation event. they bind with glucocorticoid receptors causing
exposure of nuclear localization signals [Heitzer
etal. 2007]. This results in rapid transport of the
Is the future risk of an adverse outcome or glucocorticoid receptor complex into the nucleus
response of an outcome to an intervention the which then binds to DNA altering the transcrip-
same for given eosinophil cutoffs? tion of target genes which code for pro-inflamma-
Studies have not consistently used the same spu- tory proteins thereby inhibiting the synthesis of
tum or blood eosinophil cutoff. This has led to the cytokines such as IL-3, IL-5 and GM-CSF, pro-
question of what cutoff should be applied in clini- moting eosinophil apoptosis.
cal trials and clinical practice. Importantly, differ-
ent clinical outcomes have different relationships Inhaled and oral corticosteroids decrease eosino-
with eosinophilic inflammation at a single point in philic bronchial mucosal inflammation [Djukanovi

http://taj.sagepub.com 39
Therapeutic Advances in Chronic Disease 7(1)

et al. 1992] and induced sputum differential Theophylline


eosinophil count [Brightling etal. 2000; Jatakanon Theophylline is indicated as third line with most
et al. 2000; Pizzichini et al. 1998]. Importantly, studies showing an equivalent effect and few stud-
the presence of sputum eosinophilia (>3%) in ies showing a very minimal benefit as compared
asthma and COPD [Gibson et al. 1995; Green to doubling steroid regimens or adding a long
et al. 2002a,b] and the Th2 gene signature acting beta agonist (LABA) [Lim et al. 2000;
[Woodruff et al. 2009] has shown to be a good Subramanian etal. 2015; ZuWallack etal. 2001].
predictor of the response to corticosteroid treat- Theophylline is commonly prescribed for asthma
ment in stable disease. These observations have and COPD as add on treatment in poorly con-
been extended to acute exacerbations of COPD trolled disease [Barnes, 2006; see also http://www.
with the blood eosinophil count predicting a ginasthma.org and http://www.goldcopd.org]. It
good response to oral prednisolone [Bafadhel has both bronchodilator and anti-inflammatory
etal. 2012]. Studies further showed that treatment actions with the latter possibly due to its effects
aimed to maintain eosinophil counts <23% upon the activity of a key corticosteroid-associated
decreased the number of exacerbations and corepressor protein, histone deacetylase (HDAC)2.
hospital admissions in asthma and COPD. Histone acetylation and deacetylation is key in
Interestingly in the study by Green and colleagues regulating expression of inflammatory genes
the macrophage red hue was further studied and [Barnes etal. 2005]. Theophylline is an activator
identified that in those subjects with increased red of HDAC2 even at low therapeutic levels and
hue but normal eosinophil count that corticoster- enhances the action of corticosteroids [Ito et al.
oid withdrawal was associated with onset of poor 2006]. In asthma theophylline caused significant
control and increased exacerbation risk whereas reduction in the number of eosinophils in bron-
only in those subjects without evidence of either chial biopsies, bronchoalveolar lavage (BAL) and
sputum eosinophils nor increased macrophage induced sputum [Lim et al. 2001] as compared
red hue could corticosteroids be successfully with placebo. Low-dose theophylline when used
withdrawn [Kulkarni etal. 2010]. in combination with inhaled corticosteroids sig-
nificantly decreased sputum eosinophils counts in
COPD patients as compared to theophylline alone
Leukotriene receptor antagonists [Ford etal. 2010].
Leukotrienes are potent lipid mediators derived
from arachidonic acid metabolism and released
predominately by mast cells and eosinophils. Immunomodulators
They are potent bronchoconstrictors, causing Immunomodulators such as methotrexate and
airway smooth muscle contraction and mucus cyclosporin have steroid-sparing effects, but also
hyper-secretion [Hallstrand and Henderson, predictable side-effects that have limited their
2010]. Leukotriene receptor antagonists value in severe asthma. To date only one study
decrease eosinophilic airway inflammation in has specifically investigated the effect of metho-
blood and sputum in asthma [Pizzichini et al. trexate upon peripheral blood eosinophil count
1999], albeit to a lesser extent than inhaled cor- and found no difference between the placebo or
ticosteroids [Jayaram etal. 2005]. In moderate- treatment arms [Erzurum et al. 1991]. These
to-severe disease an effect of leukotriene receptor therapies have not been tested in COPD.
antagonists upon eosinophilic airway inflamma-
tion is minimal or absent [Green et al. 2006;
Pavord etal. 2007]. In a study where montelu- Anti-IgE omalizumab
kast was used as an add on to a steroid treatment Omalizumab is the only biological therapy
regime as compared to arm where steroid dose approved for the treatment of asthma. It is a
was doubled both arms showed improvement in humanized monoclonal anti-IgE antibody. A
forced expiratory volume in 1 second (FEV1) recent Cochrane review of 25 omalizumab trials
but there was no difference in sputum eosino- in moderate-to-severe asthma showed effective-
philic inflammation with either arms [Barnes ness in decreasing exacerbations and hospitaliza-
etal. 2007]. In COPD small studies suggest lim- tions as compared with placebo with increased
ited clinical benefit and to date no studies have potential to withhold inhaled corticosteroids in
reported effects upon eosinophilic airway in- patients on anti-IgE treatment as compared with
flammation [Chauhan and Ducharme, 2012; placebo [Normansell etal. 2014]. The effects of
Ducharme etal. 2011]. omalizumab upon eosinophilic inflammation

40 http://taj.sagepub.com
L George and CE Brightling

have been studied in a single multicentre study in showed in phase II studies improvements in lung
which omalizumab reduced the sputum, epithe- function and a decrease in both sputum and
lial and sub mucosal eosinophil count [Djukanovi blood eosinophil counts [Castro et al. 2011].
et al. 2004] and high blood eosinophils, serum Improvements in asthma control were associated
periostin or FeNO prior to treatment initiation with high baseline blood eosinophil counts. Two
predict favourable responses to omalizumab duplicate phase III trials confirmed reduction in
[Hanania etal. 2013]. asthma exacerbations; improvements in health
status and asthma control [Castro etal. 2015].

Therapies in clinical development Benralizumab is a human afucosylated monoclo-


There is tremendous interest in targeting eosino- nal antibody that targets IL-5R expressed by
philic/Th2-mediated inflammation and this has led eosinophils and basophils and following receptor
to the development of both biological and small blockade initiates antibody-dependent cell-medi-
molecule therapies. Clinical trials in this space ated cytotoxicity [Ghazi et al. 2012]. A small
published since 2010 are summarized in Table 1. phase I double-blind placebo-controlled trial
showed that benralizumab decreased airway
mucosal and sputum eosinophils in addition to a
Anti-IL-5/anti-IL-5R completely suppressing blood and bone marrow
As described above, IL-5 has a pivotal role in the eosinophils and their precursors when compared
differentiation and maturation of eosinophils in with placebo [Laviolette etal. 2013]. Benralizumab
bone marrow and survival in tissue. Monoclonal showed a significant reduction in exacerbation
antibody therapies are in late phase clinical devel- rates, lung function and asthma control in severe
opment neutralising IL-5 or blocking IL-5R. asthmatics with high baseline blood eosinophil
count [Castro etal. 2014]. Although targeting the
Mepolizumab is a humanized monoclonal anti- IL-5R is likely to have more profound effects
body against free IL-5 [Desai and Brightling, upon eosinophilic inflammation than IL-5 neu-
2009; Haldar etal. 2009]. Earlier trials confirmed tralization whether this translates into differential
that anti-IL-5 monoclonal antibody therapy atten- clinical effects remains unclear.
uated markedly blood, sputum and, to a lesser
extent, bronchial mucosal eosinophils, but failed In the first study of a biologic therapy in COPD,
to demonstrate significant benefits in clinical end- benralizumab improved lung function, but did
points [Flood-Page etal. 2007; Leckie etal. 2000]. not decrease exacerbation rates, although there
Although the clinical outcomes from these studies was a trend to reduction in exacerbations and
were disappointing together they suggested a pos- improvement in health status in those COPD
sible effect upon exacerbation frequency. This subjects with higher baseline blood and sputum
gave encouragement to undertake a phase IIa eosinophil counts [Brightling etal. 2014]. These
study in severe asthmatics with evidence of eosin- encouraging findings have supported progression
ophilic inflammation and frequent exacerbations, to phase III studies.
which demonstrated a reduction in exacerbation
frequency together with reductions in eosinophilic
inflammation [Haldar et al. 2009; Pavord et al. Anti-IL-13 and IL-4R
2012]. This observation was replicated in phase The Th2-derived cytokines IL-4 and IL-13 have
IIb and III studies confirming that mepolizumab multiple effects upon inflammatory and struc-
reduced severe asthma exacerbations, together tural cells. IL-4 and IL-13 activate epithelial cells
with benefits in health status, a small increase in to release important eosinophil chemoattractants,
lung function as well as a safe glucocorticosteroids promote IgE class switching and IL-13 has direct
sparing effects [Bel etal. 2014; Ortega etal. 2014]. effects upon airway smooth muscle augmenting
Benefits were associated with the blood eosinophil hyper-contractility. IL-13 binds with IL-13R1
count and previous exacerbation frequency. with a low affinity and then with IL-4R to form
Following cessation of mepolizumab therapy ben- a high affinity cytokine binding heterodimer.
efits are lost within 6 months after treatment with- [Brightling et al. 2010]. Therefore monoclonal
drawal [Haldar etal. 2014]. antibody therapies directed towards IL-4R
inhibit the function of IL-4 in concert with IL-13
Reslizumab is another humanized monoclonal via IL-13RI. However, IL-13 neutralization has
antibody, which neutralises circulating IL-5, no effect upon IL-4, but inhibits the activation of

http://taj.sagepub.com 41
Table 1. Phase II and III parallel double-blind placebo-controlled randomized controlled trials of anti-Th2 cytokine/chemokine therapies since 2010.

Duration Intervention Subjects Primary outcomes Secondary outcomes


Corren etal. [2011] 6 months Anti-IL-13 Severe asthma, FEV1% predicted symptoms
Lebrikizumab n=219 Greater in periostinHigh Trend in exacerbation rate
Hanania [2014] 12 weeks Anti-IL-13 Severe asthma exacerbation rate FEV1% predicted
Lebrikizumab n=463 Greater in periostinHigh Greater in periostinHigh
Piper etal. [2013] 12 weeks Tralokinumab Moderate-to-severe ACQ FEV1
Anti-IL-13 asthma, 2 agonist use
n=194
Brightling etal. [2014] 52 weeks Tralokinumab Severe asthma Trend in exacerbation FEV1
Therapeutic Advances in Chronic Disease 7(1)

Anti-IL-13 n=452 rate Improved symptom scores in DPP4High


in periostinHigh DPP4High
Wenzel etal. [2013] 12 weeks Dupilumab Moderate to severe exacerbation rate FEV1
Anti IL4R asthma, Improved symptom scores
n=104
Wenzel etal. [2014] 12 weeks Dupilumab Moderate to severe ACQ -
Anti IL4R asthma
n=104
Pavord etal. [2012] 12 months Mepolizumab Severe asthma, exacerbation rate Small FEV1
Anti-IL-5 n=621 blood, sputum Small symptom scores
eosinophils
Bel etal. [2014] 20 weeks Mepolizumab Severe asthma, in glucocorticoid exacerbation rate
Anti-IL-5 n=135 daily dose
Ortega etal. [2014] 32 weeks Mepolizumab Severe asthma, exacerbation rate FEV1
Anti-IL-5 n=576 Improved symptom scores
Castro etal. [2014] 12 months Benralizumab Moderate to severe exacerbation rate. FEV1
Anti-IL-5R asthma Improved ACQ
n=609
Brightling etal. [2014] 48 weeks Benralizumab Moderate to severe exacerbation rate FEV1
Anti-IL-5R COPD Trend in exacerbation rate in blood and
n=101 sputum eosinophil group
Castro etal. [2011] 12 weeks Reslizumab Moderate to severe Improved ACQ blood, sputum eosinophils
Anti-IL5 asthma Trend in exacerbation rate
n=106
Castro etal. [2015] 12 months Reslizumab Moderate to severe exacerbation rate FEV1
Anti-IL5 asthma Improved symptom scores
n=953

(Continued)

42 http://taj.sagepub.com
Table 1. (Continued)

Duration Intervention Subjects Primary outcomes Secondary outcomes


Corren etal. [2014] 16 weeks Reslizumab Moderate to severe FEV1 greater in patients ACQ and rescue medication use in patients
Anti-IL5 asthma, with blood eosinophil with blood eosinophils >400/l
n=492 >400/l
Barnes etal. [2012] 28 days OC000459 Moderate asthma FEV1 Improved AQLQ score
CRTh2 antagonist n=107 Improved symptom
scores
Gonem etal. [2014] 12 weeks QAW039 Severe asthma in sputum eosinophils Improved AQLQ score
Dp2/CRTh2 n=61 post bronchodilator FEV1
Antagonist
Molfino etal. [2013] 6 weeks KB002 Severe asthma FEV1 symptom scores
Anti-GM-CSF n=24 in sputum eosinophils
Wenzel etal. [2014] 4 weeks ARRY 502 Mild Asthma FEV1 ACQ symptom free days
DP2 antagonist n=184
Hall etal. [2012] Six weeks BI-671800 Asthma FEV1 ACQ
DP2 antagonist n=243
Sutherland etal. 4 weeks BI-671800 Asthma FEV1 ACQ
[2012] DP2 antagonist n=388
Krug etal. [2012] 2 weeks BI-671800 Allergic rhinitis nasal symptoms, IL-4
DP2 antagonist n=146 nasal eosinophils, eotaxin levels
eosinophil activation
Fitzgerald etal. [2013] 1428 days ADC-3680 Asthma (n=24) Blocked PGD2 mediated -
DP2 antagonist Healthy (n=88) eosinophil activation by
>90%
Pettipher etal. 2014 12 weeks OC000459 Asthma FEV1 and PEF in AQLQ ACQ Exacerbation rate
DP2 antagonist n=482 eosinophilic subgroup
Busse etal. [2013] 12 weeks AMG 853 Asthma ACQ FEV1 symptom scores
D2 antagonist n=79 exacerbation
KaloBios [2014] 24 weeks KB003 Severe Asthma FEV1 ACQ exacerbations
Anti-GM-CSF n=160 FEV1 in eosinophilic
subgroup
Gaureauv etal. [2011] 9 weeks TPI ASM8 Mild Moderate sputum eosinophil Safe at all doses tested
Anti-IL-3,-4,GM- Asthma count
CSF, CCR3 n=14 sputum ECP
Greiff etal. [2010] 10 weeks AZD3778 Seasonal allergic eosinophil allergic rhinitis symptoms
CCR3 antagonist rhinitis n=38
Neighbour etal. 22 days GW76694 Asthma eosinophils FEV1 airway hyper responsiveness
[2014] CCR3 antagonist n=60 eosinophil progenitor ACQ
cell count
Abbreviations: AQLQ, asthma quality of life questionnaire; ACQ, asthma control questionnaire; COPD, chronic obstructive pulmonary disease; ECP, eosinophil cationic protein; FEV1,
forced expiratory volume in 1 second; GM-CSF, granulocyte-monocyte colony stimulating factor; IL, interleukin.

http://taj.sagepub.com 43
L George and CE Brightling
Therapeutic Advances in Chronic Disease 7(1)

IL-13RI and exerts effects upon IL-13RII which have been studied in phase II trials. BI-671800
are antibody dependent. Inhibition of IL-13RII showed better improvement in FEV1 when com-
activation occurs with tralokinumab but not leb- pared with montelukast [Hall etal. 2012], but did
rikizumab [Ultsch et al. 2013]. Increased IL-13 not show much difference in effects on FEV1
expression is a consistent feature of asthma in when compared with an inhaled steroid treat-
peripheral blood, sputum, BAL and bronchial ment. Phase II trials investigating the DP2
biopsies. IL-13 concentrations in sputum have antagonists OC000459 and QAW 039 DP2,
been seen to be related to asthma control [Saha respectively, showed a reduction in sputum eosin-
etal. 2008]. ophil counts and improved symptoms in mild
[Barnes et al. 2012] and severe asthmatics
Phase II trials have shown consistent positive [Gonem et al. 2014]. These effects were also
results for anti-IL-13 therapy [Corren etal. 2011; demonstrated with BI-671800 in seasonal allergic
Hanania, 2014; Piper etal. 2013] with improve- rhinitis [Krug etal. 2012]. A more recent phase II
ment in lung function. This effect was more pro- trial continued to show benefits of oral CRTH2
nounced in patients who had higher levels of treatment on FEV1 and symptom scores [Wenzel
pretreatment periostin, dipepityl-pepitidase-4 or etal. 2014].
evidence of increased sputum IL-13 [She et al.
2014]. Similarly in those with evidence of activa-
tion of the IL-13 pathway had lower exacerbation Anti-GM-CSF
rates in response to anti-IL13 therapy. GM-CSF causes the differentiation proliferation
and survival of monocytes, macrophages and
Anti-IL4R, dupilumab, also improves lung granulocytes including eosinophils and neutro-
function and asthma control and in an inhaled phils. The development of the anti-GM-CSF
corticosteroid withdrawal study there was signifi- IgG1 antibody MT203 showed decrease in eosin-
cant reduction in the exacerbation rates in those ophil activation and survival [Krinner etal. 2007].
subjects that received dupilumab versus placebo The safety and efficacy of other anti-GMCSF
[Teper etal. 2014; Wenzel etal. 2013]. therapies are in early clinical development and to
date whether it impacts upon eosinophilic inflam-
In the anti-IL-13 and anti-IL-4R trials the blood mation is uncertain. Phase I and II trials using
eosinophil count increased possibly due to its anti-GM-CSF treatments KB002 [Bardin et al.
indirect effects upon generation of eosinophil 2013] and KB003 [KaloBios, 2014] have been
chemokines or directly due to the effect of IL-13 done in the hope of finding a treatment to target
upon eosinophil endothelial adhesion as atopic and non-atopic asthma. These anti-GM-
described above. Interestingly whether anti- CSF therapies have shown no improvements
IL-4/13 approaches reduce sputum or bronchial compared to placebo in overall study populations,
mucosal eosinophilia is unknown. but a statistically significant improvement in
FEV1 and decrease in sputum eosinophil count
was demonstrated in the eosinophilic subgroups
Anti-CRTh2 (blood eosinophil 300cells/l).There was no
Prostaglandin D2 (PGD2) is a product of arachi- effect on symptom scores or exacerbations.
donic acid metabolism and is a major prostanoid Whether there will be further development and
found within the airways of asthmatics immedi- phase III trials of these agents to establish their
ately following an airway challenge. PGD2 is effects is uncertain at present.
known to have chemokinetic and chemotactic
effects on eosinophils and Th2 lymphocytes act-
ing via DP2/CRTh2 receptor [Hirai etal. 2001; Anti-CCR3
Nagata etal. 1999]. This interaction plays a key The CCR3 chemokine receptor is expressed on
role in airway hyper-responsiveness, IgE and the surface of eosinophils and is activated by
cytokine production [Pettipher and Whittaker, chemokines Eotaxin 1 (CCL11)/Eotaxin 2 (CCL
2012]. An in vitro study using a potent CRTH2 24) and Eotaxin 3(CCL26). Eotaxin 1(CCL11)
antagonist was the first to show decrease in has been shown to be actively involved in eosino-
PGD2-mediated human eosinophil migration phil recruitment and in survival along with IL-5.
[Sugimoto et al. 2003]. Since this discovery a Airway epithelial cells and airway smooth muscle
variety of DP2/CRTH2 antagonists including cells express CCR3 and also produce cytokines
ARRY 502, BI-671800, QAW-039, OC000459 and fibroblast growth factor. Recent studies using

44 http://taj.sagepub.com
L George and CE Brightling

CCR3 antagonists including GSK766994, GSK our understanding of the role of the eosinophil in
766904,GW824575, DPC 168 and QAP 642 asthma and COPD and sharpen our focus on the
[Pease and Horuk, 2014] in asthma or in allergic need for precision medicine.
rhinitis have not demonstrated significant effects
upon eosinophilic airway inflammation and have Funding
questioned the importance of CCR3 in eosino- The authors received no financial support for the
phil recruitment versus other mechanisms such as research, authorship, and/or publication of this
CRTh2 [Neighbour etal. 2014]. article.

Declaration of conflicting interest


Conclusion CEB has received grant and consultancy funds
Current treatments and the emergence of new via his Institution from AstraZeneca/MedImmune,
therapies targeting eosinophilic inflammation have GSK, Roche/Genentech, Novartis, Chiesi and
informed our understanding of the relationship Boehringer-Ingelheim.
between eosinophilic inflammation and different
characteristics of asthma and COPD. Identification
of eosinophilic and Th2-meduiated inflammation References
predicts a good response to corticosteroid, anti- Amelink, M., Nijs, S., Groot, J., Tilburg, P., Spiegel,
IgE, anti-IL5, anti-IL5R, and anti-13 therapies. P., Krouwels, F. etal. (2013) Three phenotypes of
Eosinophilic inflammation has a clear role in adult-onset asthma. Allergy 68: 674680.
asthma exacerbations and its inhibition by anti- Ayars, G., Altman, L., Gleich, G., Loegering, D. and
IL5 improves lung function, symptoms and health Baker, C. (1985) Eosinophil-and eosinophil granule-
status related to the intensity of eosinophilic mediated pneumocyte injury. J Allergy Clin Immunol
inflammation and possibly with greater effects 76: 595604.
upon lung function following IL-5R blockade Bafadhel, M., McKenna, S., Terry, S., Mistry,
rather than IL-5 neutralization. In COPD, anti- V., Pancholi, M., Venge, P. etal. (2012) Blood
IL-5 improved lung function in eosinophilic eosinophils to direct corticosteroid treatment of
COPD, but the role of eosinophils in COPD exac- exacerbations of chronic obstructive pulmonary
erbations remains uncertain. Anti-IL-13 has con- disease: a randomized placebo-controlled trial. Am J
sistent effects upon lung function in asthma, but Respir Crit Care Med 186: 4855.
the benefits in exacerbations have to date been less Bafadhel, M., McKenna, S., Terry, S., Mistry, V.,
marked than with anti-IL-5 and current late phase Reid, C., Haldar, P. etal. (2011) Acute exacerbations
studies in selected populations will be critical in of chronic obstructive pulmonary disease:
determining the role of IL-13 in asthma exacerba- identification of biologic clusters and their biomarkers.
tions. CCR3 antagonism has been disappointing Am J Respir Crit Care Med 184: 662671.
whereas anti-CRTh2 is emerging as a potentially Bardin, P., Thompson, P., Luckey, A., Yarranton,
important target that impacts inflammation, lung G. and Molfino, N. (2013) A randomized placebo-
function and symptoms with effects upon exacer- controlled safety and pharmacodynamic study
bations to be determined. The potential role of of KB002, a chimeric antiGMCSF monoclonal
therapies targeting IL-4/13 and CRTh2 in eosino- antibody, in patients with asthma. Am J Respir Crit
philic COPD is unknown. Care Med 187: A3867.
Barnes, P. (2006) Theophylline for COPD. Thorax
In the forthcoming 5 years the mechanistic role of 61: 742744.
the eosinophil in asthma and COPD, the propor-
tion of the asthma and COPD patients in whom Barnes, P., Adcock, I. and Ito, K. (2005) Histone
the eosinophil is critical and the magnitude of acetylation and deacetylation: importance in
inflammatory lung diseases. Eur Respir J 25: 552563.
clinical efficacy of anti-eosinophil therapies will
become clear. During this time attention will Barnes, N., Laviolette, M., Allen, D., Flood-Page,
need to focus on the aspects of disease that are P., Hargreave, F., Corris, P. etal. (2007) Effects of
independent of eosinophilic inflammation and in montelukast compared to double dose budesonide on
those where the eosinophil plays a key role then airway inflammation and asthma control. Respir Med
there is a need to understand the relative merits of 101: 16521658.
each therapy in an individual and how can we Barnes, N., Pavord, I., Chuchalin, A., Bell, J.,
apply biomarkers to predict long term outcomes. Hunter, M., Lewis, T. etal. (2012) A randomized,
Addressing this challenge will both further inform double-blind, placebo-controlled study of the CRTH2

http://taj.sagepub.com 45
Therapeutic Advances in Chronic Disease 7(1)

antagonist OC000459 in moderate persistent asthma. Busse, W., Wenzel, S., Meltzer, E., Kerwin, E.,
Clin Exp Allergy 42: 3848. Liu, M., Zhang, N. etal. (2013) Safety and efficacy
of prostaglandin D2 receptor antagonist AMG 853
Bel, E., Ortega, H. and Pavord, I. (2014) in asthmatic patients. J Allergy Clin Immunol 131:
Glucocorticoids and mepolizumab in eosinophilic 339345.
asthma. N Engl J Med 371: 2434.
Castro, M., Mathur, S., Hargreave, F., Boulet, L.,
Berry, M., Shaw, D., Green, R., Brightling, C., Xie, F., Young, J. etal. (2011) Reslizumab for poorly
Wardlaw, A. and Pavord, I. (2005) The use of exhaled controlled, eosinophilic asthma: a randomized,
nitric oxide concentration to identify eosinophilic placebo-controlled study. Am J Respir Crit Care Med
airway inflammation: an observational study in adults 184: 11251132.
with asthma. Clin Exp Allergy 35: 11751179.
Castro, M., Wenzel, S., Bleecker, E., Pizzichini, E.,
Boorsma, M., Lutter, R., van de Pol, M., Out, T., Kuna, P., Busse, W. etal. (2014) Benralizumab, an
Jansen, H. and Jonkers, R. (2007) Repeatability anti-interleukin 5 receptor monoclonal antibody,
of inflammatory parameters in induced sputum of versus placebo for uncontrolled eosinophilic asthma:
COPD patients. J Chron Obstruct Pulmon Dis 4: a phase 2b randomised dose-ranging study. Lancet
321329. Respir Med 2: 879890.
Brightling, C. (2013) Biomarkers that predict and Castro, M., Zangrilli, J., Wechsler, M., Bateman, E.,
guide therapy for exacerbations of chronic obstructive Brusselle, G., Bardin, P. etal. (2015) Reslizumab for
pulmonary disease. Ann Am Thorac Soc 10: inadequately controlled asthma with elevated blood
S214S219. eosinophil counts: results from two multicentre,
Brightling, C., Bleecker, E., Panettieri, R., Bafadhel, parallel, double-blind, randomised, placebo-controlled,
M., She, D., Ward, C. etal. (2014) Benralizumab for phase 3 trials. Lancet Respir Med 3: 355366.
chronic obstructive pulmonary disease and sputum Chauhan, B. and Ducharme, F. (2012) Anti-
eosinophilia: a randomised, double-blind, placebo- leukotriene agents compared to inhaled corticosteroids
controlled, phase 2a study. Lancet Respir Med 2: in the management of recurrent and/or chronic
891901. asthma in adults and children. Cochrane Database Syst
Brightling, C., Bradding, P., Symon, F., Holgate, Rev 5: CD002314.
S., Wardlaw, A. and Pavord, I. (2002) Mast-cell Corren, J., Lemanske, R., Jr., Hanania, N., Korenblat,
infiltration of airway smooth muscle in asthma. N Engl P., Parsey, M., Arron, J. etal. (2011) Lebrikizumab
J Med 346: 16991705. treatment in adults with asthma. N Engl J Med 365:
Brightling, C., McKenna, S., Hargadon, B., 10881098.
Birring, S., Green, R., Siva, R. etal. (2005) Sputum Corren, J., Weinstein, J., Janka, L., OBrien, C. and
eosinophilia and the short term response to inhaled Zangrilli, J. (2014) A randomised phase 3 study of
mometasone in chronic obstructive pulmonary reslizumab efficacy in relation to blood eosinophil
disease. Thorax 60: 193198. levels in patients with moderate to severe asthma. Eur
Brightling, C., Monteiro, W., Ward, R., Parker, Respir J 44(Suppl. 58): 4673.
D., Morgan, M., Wardlaw, A. etal. (2000) Sputum Davoine, F. and Lacy, P. (2014) Eosinophil cytokines,
eosinophilia and short-term response to prednisolone chemokines, and growth factors: emerging roles in
in chronic obstructive pulmonary disease: a immunity. Front Immunol 5: 570.
randomised controlled trial. Lancet 356: 14801485.
Denburg, J. (1998) The origins of basophils and
Brightling, C., Saha, S. and Hollins, F. (2010) eosinophils in allergic inflammation. J Allergy Clin
Interleukin-13: prospects for new treatments. Clin Exp Immunol 102: S74S76.
Allergy 40: 4249.
Desai, D. and Brightling, C. (2009) Cytokine and
Brightling, C., Symon, F., Birring, S., Bradding, P., anti-cytokine therapy in asthma: ready for the clinic?
Wardlaw, A. and Pavord, I. (2003) Comparison of Clin Exp Immunol 158: 1019.
airway immunopathology of eosinophilic bronchitis
and asthma. Thorax 58: 528532. Desai, D., Newby, C., Symon, F., Haldar, P.,
Shah, S., Gupta, S. etal. (2013) Elevated sputum
Brown, H. (1958) Treatment of chronic asthma interleukin-5 and submucosal eosinophilia in obese
with prednisolone significance of eosinophils in the individuals with severe asthma. Am J Respir Crit Care
sputum. Lancet 272: 12451247. Med 188: 657663.
Burrowes, K., Doel, T. and Brightling, C. (2014) Djukanovi, R., Wilson, J., Britten, K., Wilson, S.,
Computational modeling of the obstructive lung Walls, A., Roche, W. etal. (1992) Effect of an inhaled
diseases asthma and COPD. J Transl Med 12(Suppl. corticosteroid on airway inflammation and symptoms
2): S5. in asthma. Am Rev Respir Dis 145: 669674.

46 http://taj.sagepub.com
L George and CE Brightling

Djukanovi, R., Wilson, S., Kraft, M., Jarjour, N., Frigas, E., Motojima, S. and Gleich, G. (1991) The
Steel, M., Chung, K. etal. (2004) Effects of treatment eosinophilic injury to the mucosa of the airways in the
with anti-immunoglobulin E antibody omalizumab on pathogenesis of bronchial asthma. Eur Respir J Suppl
airway inflammation in allergic asthma. Am J Respir 13: 123s135s.
Crit Care Med 170: 583593.
Fukuda, T., Fukushima, Y., Numao, T., Ando,
Douwes, J., Gibson, P., Pekkanen, J. and Pearce, N. N., Arima, M., Nakajima, H. etal. (1996) Role of
(2002) Non-eosinophilic asthma: importance and interleukin-4 and vascular cell adhesion molecule-1
possible mechanisms. Thorax 57: 643648. in selective eosinophil migration into the airways in
allergic asthma. Am J Respir Cell Mol Biol 14: 8494.
Ducharme, F., Lasserson, T. and Cates, C. (2011)
Addition to inhaled corticosteroids of long-acting Fulkerson, P. and Rothenberg, M. (2013) Targeting
beta2-agonists versus anti-leukotrienes for chronic eosinophils in allergy, inflammation and beyond. Nat
asthma. Cochrane Database Syst Rev 5: CD003137. Rev Drug Discov 12: 117129.
Edwards, S., Von Maltzahn, R., Naya, I. and Harrison, Gauvreau, G., Pageau, R., Sguin, R., Carballo, D.,
T. (2010) Budesonide/formoterol for maintenance and Gauthier, J., DAnjou, H. etal. (2011)Dose response
reliever therapy of asthma: a meta analysis of randomised effects of TPI ASM8 in asthmatic after allergen.
controlled trials. Int J Clin Pract 64: 619627. Allergy 66: 12421248.
Eltboli, O., Bafadhel, M., Hollins, F., Wright, A., Ghazi, A., Trikha, A. and Calhoun, W. (2012)
Hargadon, B., Kulkarni, N. etal. (2014) COPD Benralizumaba humanized mAb to IL-5 with
exacerbation severity and frequency is associated with enhanced antibody-dependent cell-mediated
impaired macrophage efferocytosis of eosinophils. cytotoxicitya novel approach for the treatment of
BMC Pulm Med 14: 112. asthma. Expert Opin Biol Ther 12: 113118.
Eltboli, O. and Brightling, C. (2013) Eosinophils as Ghebre, M., Bafadhel, M., Desai, D., Cohen, S.,
diagnostic tools in chronic lung disease. Expert Rev Newbold, P., Rapley, L. etal. (2015) Biological
Respir Med 7: 3342. clustering supports both Dutch and British
hypotheses of asthma and chronic obstructive
Eltboli, O., Mistry, V., Barker, B. and Brightling, C.
pulmonary disease. J Allergy Clin Immunol 135: 6372.
(2015) Relationship between blood and bronchial
submucosal eosinophilia and reticular basement Gibson, P., Denburg, J., Dolovich, J., Ramsdale, E.
membrane thickening in chronic obstructive and Hargreave, F. (1989) Chronic cough: eosinophilic
pulmonary disease. Respirology 20: 667670. bronchitis without asthma. Lancet 333: 13461348.
Erzurum, S., Leff, J., Cochran, J., Ackerson, L., Gibson, P., Hargreave, F., Girgis-Gabardo, A., Morris,
Szefler, S., Martin, R. etal. (1991) Lack of benefit of M., Denburg, J. and Dolovich, J. (1995) Chronic
methotrexate in severe, steroid-dependent asthma: a cough with eosinophilic bronchitis: examination
double-blind, placebo-controlled study. Ann Intern for variable airflow obstruction and response to
Med 114: 353360. corticosteroid. Clin Exp Allergy 25: 127132.
Fitzgerald, M., Snape, S., Febbraro, S., Banyard, A., Gonem, S., Berair, R., Singapuri, A., Hartley, R.,
Patnell, C., Ruparelia, B. etal. (2013) The safety, PK, Laurencin, M., Bacher, G. etal. (2014) Late-breaking
PD profile of AD3680, a potent and selective CRTH2 abstract: phase 2a randomized placebo-controlled trial
antagonist in healthy volunteers and partly controlled of the oral prostaglandin D2 receptor (DP2/CRTh2)
atopic asthmatic subjects. C-23 Novel therapeutic in antagonist QAW039 in eosinophilic asthma. Eur
asthma. ATS Conference Proceedings, Pennsylvania, 21 Respir J 44(Suppl. 58): 2908.
May 2013. Abstract A3874.
Green, R., Brightling, C., McKenna, S., Hargadon,
Flood-Page, P., Menzies-Gow, A., Phipps, S., Ying, B., Neale, N., Parker, D. etal. (2006) Comparison
S., Wangoo, A., Ludwig, M. etal. (2003) Anti-IL-5 of asthma treatment given in addition to inhaled
treatment reduces deposition of ECM proteins in the corticosteroids on airway inflammation and
bronchial subepithelial basement membrane of mild responsiveness. Eur Respir J 27: 11441151.
atopic asthmatics. J Clin Invest 112: 10291036.
Green, R., Brightling, C., McKenna, S., Hargadon,
Flood-Page, P., Swenson, C., Faiferman, I., B., Parker, D., Bradding, P. etal. (2002a) Asthma
Matthews, J., Williams, M., Brannick, L. etal. exacerbations and sputum eosinophil counts: a
(2007) A study to evaluate safety and efficacy of randomised controlled trial. Lancet 360: 17151721.
mepolizumab in patients with moderate persistent
Green, R., Brightling, C., Woltmann, G., Parker,
asthma. Am J Respir Crit Care Med 176: 10621071.
D., Wardlaw, A. and Pavord, I. (2002b) Analysis of
Ford, P., Durham, A., Russell, R., Gordon, F., induced sputum in adults with asthma: identification
Adcock, I. and Barnes, P. (2010) Treatment effects of subgroup with isolated sputum neutrophilia and
of low-dose theophylline combined with an inhaled poor response to inhaled corticosteroids. Thorax 57:
corticosteroid in COPD. Chest 137: 13381344. 875879.

http://taj.sagepub.com 47
Therapeutic Advances in Chronic Disease 7(1)

Greiff, L., Ahlstrm-Emanuelsson, C., Bahl, A., Jatakanon, A., Lim, S. and Barnes, P. (2000) Changes
Bengtsson, T., Dahlstrm, K., Erjeflt, J. etal. in sputum eosinophils predict loss of asthma control.
(2010) Effects of dual CCR3 and H-1 antagnonist on Am J Respir Crit Care Med 161: 6472.
symptoms and eosinophilic inflammation in allergic
Jayaram, L., Pizzichini, M., Cook, R., Boulet, L.,
rhinitis. Respir Res 11: 17.
Lemiere, C., Pizzichini, E. etal. (2006) Determining
Haldar, P., Brightling, C., Hargadon, B., Gupta, S., asthma treatment by monitoring sputum cell counts:
Monteiro, W., Sousa, A. etal. (2009) Mepolizumab effect on exacerbations. Eur Respir J 27: 483494.
and exacerbations of refractory eosinophilic asthma.
Jayaram, L., Pizzichini, E., Lemiere, C., Man, S.,
N Engl J Med 360: 973984.
Cartier, A., Hargreave, F. etal. (2005) Steroid naive
Haldar, P., Brightling, C., Singapuri, A., Hargadon, eosinophilic asthma: anti-inflammatory effects of
B., Gupta, S., Monteiro, W. etal. (2014) Outcomes fluticasone and montelukast. Thorax 60: 100105.
after cessation of mepolizumab therapy in severe
Jia, G., Erickson, R., Choy, D., Mosesova, S., Wu,
eosinophilic asthma: a 12-month follow-up analysis.
L., Solberg, O. etal. (2012) Periostin is a systemic
J Allergy Clin Immunol 133: 921.
biomarker of eosinophilic airway inflammation in
Haldar, P., Pavord, I., Shaw, D., Berry, M., Thomas, asthmatic patients. J Allergy Clin Immunol 130: 647654.
M., Brightling, C. etal. (2008) Cluster analysis and KaloBios (2014) KaloBios Reports Top-Line Data
clinical asthma phenotypes. Am J Respir Crit Care Med for Phase 2 Study of KB003 in Severe Asthma.
178: 218224. Available at: http://ir.kalobios.com/releasedetail.
Hall, I., Sarno, M., Disse, B., Atis, S., Bateman, cfm?ReleaseID=821931 (accessed July 2015).
E. and Finnigan, H. (2012) Efficacy and safety Kay, A. (2014) The early history of the eosinophil.
of BI 671800, an oral CRTH2 antagonist, as add Clin Exp Allergy 45: 575582.
on therapy in poorly controlled asthma patients
prescribed an inhaled corticosteroid. Eur Respir J Kay, A., Phipps, S. and Robinson, D. (2004) A role
40(Suppl. 56): abstract 2551. for eosinophils in airway remodelling in asthma.
Trends Immunol 25: 477482.
Hallstrand, T. and Henderson, W., Jr. (2010) An
update on the role of leukotrienes in asthma. Curr Keatings, V., Evans, D., OConnor, B. and Barnes,
Opin Allergy Clin Immunol 10: 6066. P. (1997) Cellular profiles in asthmatic airways: a
comparison of induced sputum, bronchial washings,
Hanania, N. (2014) Efficacy and safety of lebrikizumab and bronchoalveolar lavage fluid. Thorax 52: 372374.
in severe uncontrolled asthma: results from the lute
and verse phase II randomized, double-blind, placebo- Krinner, E., Raum, T., Petsch, S., Bruckmaier, S.,
controlled trials. J Allergy Clin Immunol 133: AB402. Schuster, I., Petersen, L. etal. (2007) A human
monoclonal IgG1 potently neutralizing the pro-
Hanania, N., Wenzel, S., Rosn, K., Hsieh, H., inflammatory cytokine GM-CSF. Mol Immunol 44:
Mosesova, S., Choy, D. etal. (2013) Exploring the 916925.
effects of omalizumab in allergic asthma: an analysis
of biomarkers in the EXTRA study. Am J Respir Crit Krug, N., Gupta, A., Badorrek, D., Mueller, M.,
Care Med 187: 804811. Casper, A., Pivovarova, A. etal. (2012) CRTH2
antagonist, BI 671800 (BI), reduces nasal symptoms and
Heitzer, M., Wolf, I., Sanchez, E., Witchel, S. inhibits nasal cytokines and eosinophils in SAR patients
and DeFranco, D. (2007) Glucocorticoid receptor exposed to grass pollen in an environmental challenge
physiology. Rev Endocr Metab Disord 8: 321330. chamber (ECC). Am J Resp Crit Care Med 185: A4185.
Hirai, H., Tanaka, K., Yoshie, O., Ogawa, Kulkarni, N., Hollins, F., Sutcliffe, A., Saunders,
K., Kenmotsu, K., Takamori, Y. etal. (2001) R., Shah, S., Siddiqui, S. etal. (2010) Eosinophil
Prostaglandin D2 selectively induces chemotaxis in protein in airway macrophages: a novel biomarker of
T helper type 2 cells, eosinophils, and basophils via eosinophilic inflammation in patients with asthma.
seven-transmembrane receptor CRTH2. J Exp Med J Allergy Clin Immunol 126: 6169.
193: 255261.
Laviolette, M., Gossage, D., Gauvreau, G., Leigh,
Humbles, A., Lloyd, C., McMillan, S., Friend, D., R., Olivenstein, R., Katial, R. etal. (2013) Effects
Xanthou, G., McKenna, E. etal. (2004) A critical role of benralizumab on airway eosinophils in asthmatic
for eosinophils in allergic airways remodeling. Science patients with sputum eosinophilia. J Allergy Clin
305: 17761779. Immunol 132: 10861096.
Ito, K., Yamamura, S., Essilfie-Quaye, S., Cosio, B., Leckie, M., Brinke, A., Khan, J., Diamant, Z.,
Ito, M., Barnes, P. etal. (2006) Histone deacetylase OConnor, B., Walls, C. etal. (2000) Effects of
2-mediated deacetylation of the glucocorticoid an interleukin-5 blocking monoclonal antibody on
receptor enables NF-kappaB suppression. J Exp Med eosinophils, airway hyper-responsiveness, and the late
203: 713. asthmatic response. Lancet 356: 21442148.

48 http://taj.sagepub.com
L George and CE Brightling

Leigh, R., Pizzichini, M., Morris, M., Maltais, Moqbel, R., Levi-Schaffer, F. and Kay, A. (1994)
F., Hargreave, F. and Pizzichini, E. (2006) Stable Cytokine generation by eosinophils. J Allergy Clin
COPD: predicting benefit from high-dose inhaled Immunol 94: 11831188.
corticosteroid treatment. Eur Respir J 27:
Mutalithas, K., Guillen, C., Day, C., Brightling,
964971.
C., Pavord, I. and Wardlaw, A. (2010) CRTH2
Lewis, R., Soter, N., Diamond, P., Austen, K., Oates, expression on T cells in asthma. Clin Exp Immunol
J. and Roberts, L., 2nd. (1982) Prostaglandin D2 161: 3440.
generation after activation of rat and human mast cells
Nagata, K., Hirai, H., Tanaka, K., Ogawa, K., Aso,
with anti-IgE. J Immunol 129: 16271631.
T., Sugamura, K. etal. (1999) CRTH2, an orphan
Lim, S., Jatakanon, A., Gordon, D., Macdonald, C., receptor of T-helper-2-cells, is expressed on basophils
Chung, K. and Barnes, P. (2000) Comparison of high and eosinophils and responds to mast cell-derived
dose inhaled steroids, low dose inhaled steroids plus factor (s). FEBS Lett 459: 195199.
low dose theophylline, and low dose inhaled steroids
Navarro, S., Aleu, J., Jimenez, M., Boix, E.,
alone in chronic asthma in general practice. Thorax
Cuchillo, C. and Nogues, M. (2008) The cytotoxicity
55: 837841.
of eosinophil cationic protein/ribonuclease 3
Lim, S., Tomita, K., Carramori, G., Jatakanon, on eukaryotic cell lines takes place through its
A., Oliver, B., Keller, A. etal. (2001) Low-dose aggregation on the cell membrane. Cell Mol Life Sci
theophylline reduces eosinophilic inflammation but 65: 324337.
not exhaled nitric oxide in mild asthma. Am J Respir Neighbour, H., Boulet, L., Lemiere, C., Sehmi, R.,
Crit Care Med 164: 273276. Leigh, R., Sousa, A. etal. (2014) Safety and efficacy
Loymans, R., Gemperli, A., Cohen, J., Rubinstein, of an oral CCR3 antagonist in patients with asthma
S., Sterk, P., Reddel, H. etal. (2014) Comparative and eosinophilic bronchitis: a randomized, placebo-
effectiveness of long term drug treatment strategies to controlled clinical trial. Clin Exp Allergy 44: 508516.
prevent asthma exacerbations: network meta-analysis. Newby, C., Agbetile, J., Hargadon, B., Monteiro, W.,
BMJ 348: g3009. Green, R., Pavord, I. etal. (2014a) Lung function
Luijk, B., Lindemans, C., Kanters, D., van der decline and variable airway inflammatory pattern:
Heijde, R., Bertics, P., Lammers, J. etal. (2005) longitudinal analysis of severe asthma. J Allergy Clin
Gradual increase in priming of human eosinophils Immunol 134: 287294.
during extravasation from peripheral blood to the Newby, C., Heaney, L., Menzies-Gow, A., Niven,
airways in response to allergen challenge. J Allergy R., Mansur, A., Bucknall, C. etal. (2014b) Statistical
Clin Immunol 115: 9971003. cluster analysis of the British Thoracic Society Severe
Maestrelli, P., Saetta, M., Di Stefano, A., Calcagni, refractory Asthma Registry: clinical outcomes and
P., Turato, G., Ruggieri, M. etal. (1995) Comparison phenotype stability. PLoS One 9: e102987.
of leukocyte counts in sputum, bronchial biopsies, and Normansell, R., Walker, S., Milan, S., Walters, E. and
bronchoalveolar lavage. Am J Respir Crit Care Med Nair, P. (2014) Omalizumab for asthma in adults and
152: 19261931. children. Cochrane Database Syst Rev 1: CD003559.
McGrath, K., Icitovic, N., Boushey, H., Lazarus, S., Ortega, H., Liu, M., Pavord, I., Brusselle, G.,
Sutherland, E., Chinchilli, V. etal. (2012) A large FitzGerald, J., Chetta, A. etal. (2014) Mepolizumab
subgroup of mild-to-moderate asthma is persistently treatment in patients with severe eosinophilic asthma.
noneosinophilic. Am J Respir Crit Care Med 185: N Engl J Med 371: 11981207.
612619.
Patel, K. (1998) Eosinophil tethering to interleukin-4-
Mesquita-Santos, F., Vieira-de-Abreu, A., Calheiros, activated endothelial cells requires both P-selectin and
A., Figueiredo, I., Castro-Faria-Neto, H., Weller, P. vascular cell adhesion molecule-1. Blood 92: 39043911.
etal. (2006) Cutting edge: prostaglandin D2
Pavord, I., Korn, S., Howarth, P., Bleecker, E.,
enhances leukotriene C4 synthesis by eosinophils
Buhl, R., Keene, O. etal. (2012) Mepolizumab for
during allergic inflammation: synergistic in vivo
severe eosinophilic asthma (DREAM): a multicentre,
role of endogenous eotaxin. J Immunol 176:
double-blind, placebo-controlled trial. Lancet 380:
13261330.
651659.
Molfino, N., Bardin, P., Thompson, P., Lucky, A.
Pavord, I., Pizzichini, M., Pizzichini, E. and
and Yarranton, G. (2013) A randomised placebo
Hargreave, F. (1997) The use of induced sputum to
controlled safety and pharmacodynamic study of
investigate airway inflammation. Thorax 52: 498501.
KB002, a Chimeric anti GM-CSF monoclonal
antibody, in patients with asthma. Am J Respir Crit Pavord, I., Woodcock, A., Parker, D., Rice, L.
Care Med 187: A3867. and SOLTA Study Group. (2007) Salmeterol plus

http://taj.sagepub.com 49
Therapeutic Advances in Chronic Disease 7(1)

fluticasone propionate versus fluticasone propionate exacerbations. Am J Respir Crit Care Med 150:
plus montelukast: a randomised controlled trial 16461652.
investigating the effects on airway inflammation in
Saha, S., Berry, M., Parker, D., Siddiqui, S., Morgan,
asthma. Respir Res 8: 67.
A., May, R. etal. (2008) Increased sputum and
Pease, J. and Horuk, R. (2014) Recent progress in bronchial biopsy IL-13 expression in severe asthma.
the development of antagonists to the chemokine J Allergy Clin Immunol 121: 685691.
receptors CCR3 and CCR4. Expert Opin Drug Discov
Sakula, A. (1986) CharcotLeyden crystals and
9: 467483.
Curschmann spirals in asthmatic sputum. Thorax 41:
Pettipher, R., Perkins, M., Collins, L., Baillet, M., 503507.
Lewis, T., Steiner, J. etal. (2014) The potent &
selective CRTH2 antagonist OC000459 is effective in Schleich, F., Manise, M., Sele, J., Henket, M., Seidel,
the treatment of eosinophilic asthma when given once L. and Louis, R. (2013) Distribution of sputum
daily. J Allergy Clin Immunol 133: AB3. cellular phenotype in a large asthma cohort: predicting
factors for eosinophilic vs neutrophilic inflammation.
Pettipher, R. and Whittaker, M. (2012) Update on BMC Pulm Med 13: 11.
the development of antagonists of chemoattractant
receptor-homologous molecule expressed on Th2 cells Sethi, S. and Murphy, T. (2008) Infection in the
(CRTH2). From lead optimization to clinical proof- pathogenesis and course of chronic obstructive
of-concept in asthma and allergic rhinitis. J Med Chem pulmonary disease. N Engl J Med 359: 23552365.
55: 29152931. She, D., Ranade, K., Piper, E. and Brightling, C.
Piper, E., Brightling, C., Niven, R., Oh, C., Faggioni, (2014) Efficacy and safety Of tralokinumab, an
R., Poon, K. etal. (2013) A phase II placebo- anti-Il-13 monoclonal antibody, in a phase 2b study
controlled study of tralokinumab in moderate-to- of uncontrolled severe Asthma. Am J Respir Crit Care
severe asthma. Eur Respir J 41: 330338. Med 189: A6670.

Pizzichini, E., Leff, J., Reiss, T., Hendeles, L., Boulet, Siddiqui, S., Mistry, V., Doe, C., Roach, K.,
L., Wei, L. etal. (1999) Montelukast reduces airway Morgan, A., Wardlaw, A. etal. (2008) Airway
eosinophilic inflammation in asthma: a randomized, hyperresponsiveness is dissociated from airway wall
controlled trial. Eur Respir J 14: 1218. structural remodeling. J Allergy Clin Immunol 122:
335341.
Pizzichini, E., Pizzichini, M., Efthimiadis, A., Evans, S.,
Morris, M., Squillace, D. etal. (1996) Indices of airway Singh, D., Kolsum, U., Brightling, C., Locantore, N.,
inflammation in induced sputum: reproducibility and Agusti, A., Tal-Singer, R. etal. (2014) Eosinophilic
validity of cell and fluid-phase measurements. Am J inflammation in COPD: prevalence and clinical
Respir Crit Care Med 154: 308317. characteristics. Eur Respir J 44: 16971700.

Pizzichini, E., Pizzichini, M., Gibson, P., Siva, R., Green, R., Brightling, C., Shelley, M.,
Parameswaran, K., Gleich, G., Berman, L. etal. Hargadon, B., McKenna, S. etal. (2007) Eosinophilic
(1998) Sputum eosinophilia predicts benefit from airway inflammation and exacerbations of COPD: a
prednisone in smokers with chronic obstructive randomised controlled trial. Eur Respir J 29: 906913.
bronchitis. Am J Respir Crit Care Med 158: 15111517. Smit, J. and Lukacs, N. (2006) A closer look at
Reddel, H., Taylor, D., Bateman, E., Boulet, L., chemokines and their role in asthmatic responses. Eur
Boushey, H., Busse, W. etal. (2009) An official J Pharmacol 533: 277288.
American Thoracic Society/European Respiratory Stanescu, D., Sanna, A., Veriter, C., Kostianev,
Society statement: asthma control and exacerbations: S., Calcagni, P., Fabbri, L. etal. (1996) Airways
standardizing endpoints for clinical asthma trials and obstruction, chronic expectoration, and rapid decline
clinical practice. Am J Respir Crit Care Med 180: 5999. of FEV1 in smokers are associated with increased
Rosenberg, H., Phipps, S. and Foster, P. (2007) levels of sputum neutrophils. Thorax 51: 267271.
Eosinophil trafficking in allergy and asthma. J Allergy Subramanian, R., Jindal, A., Viswambhar, V. and
Clin Immunol 119: 13031310. Babu, A. (2015) The study of efficacy, tolerability and
Rutgers, S., Timens, W., Kaufmann, H., van der safety of theophylline given along with formoterol plus
Mark, T., Koeter, G. and Postma, D. (2000) budesonide in COPD. J Clin Diagn Res 9: OC10OC13.
Comparison of induced sputum with bronchial wash,
Sugimoto, H., Shichijo, M., Iino, T., Manabe, Y.,
bronchoalveolar lavage and bronchial biopsies in
Watanabe, A., Shimazaki, M. etal. (2003) An orally
COPD. Eur Respir J 15: 109115.
bioavailable small molecule antagonist of CRTH2,
Saetta, M., Di Stefano, A., Maestrelli, P., Turato, ramatroban (BAY u3405), inhibits prostaglandin
G., Ruggieri, M., Roggeri, A. etal. (1994) D2-induced eosinophil migration in vitro. J Pharmacol
Airway eosinophilia in chronic bronchitis during Exp Ther 305: 347352.

50 http://taj.sagepub.com
L George and CE Brightling

Sutherland, R., Tetzlaff, K., Nivens, C., Tsukayama, Wenzel, S., Hopkins, R., Saunders, M., Chantry,
M., Guerreros, A., Manuel, R. etal. (2012) Efficacy D., Anderson, L., Aitchison, R. etal. (2014) Safety
and safety of BI671800, an oral CRTH2 antagnoist and efficacy of ARRY-502, a potent, selective, oral
in controller nave patients with poorly controlled CRTh2 antagonist, in patients with mild to moderate
asthma. Eur Respir J 40(Suppl. 56): 3084. Th2-driven asthma. J Allergy Clin Immunol 133: AB4.
Sweeney, J., Brightling, C., Menzies-Gow, A., Niven, Woltmann, G., McNulty, C., Dewson, G.,
R., Patterson, C., Heaney, L. etal. (2012) Clinical Symon, F. and Wardlaw, A. (2000) Interleukin-13
management and outcome of refractory asthma in induces PSGL-1/P-selectin-dependent adhesion of
the UK from the British Thoracic Society Difficult eosinophils, but not neutrophils, to human umbilical
Asthma Registry. Thorax 67: 754756. vein endothelial cells under flow. Blood 95:
31463152.
Symon, F., Lawrence, M., Williamson, M., Walsh,
G., Watson, S. and Wardlaw, A. (1996) Functional Woodruff, P., Boushey, H., Dolganov, G., Barker,
and structural characterization of the eosinophil C., Yang, Y., Donnelly, S. etal. (2007) Genome-wide
P-selectin ligand. J Immunol 157: 17111719. profiling identifies epithelial cell genes associated
with asthma and with treatment response to
Teper, A., Wang, L., Pirozzi, G., Radin, A., Graham, corticosteroids. Proc Natl Acad Sci USA 104:
N., Castro, M. etal. (2014) ACQ5 improvement with 1585815863.
dupilumab in patients with persistent asthma and
elevated eosinophil levels: responder analysis from a Woodruff, P., Modrek, B., Choy, D., Jia, G., Abbas,
12-week proof-of-concept placebo-controlled trial. Am A., Ellwanger, A. etal. (2009) T-helper type 2driven
J Respir Crit Care Med 189: A1323. inflammation defines major subphenotypes of asthma.
Am J Respir Crit Care Med 180: 388395.
Ultsch, M., Bevers, J., Nakamura, G., Vandlen,
R., Kelley, R., Wu, L. etal. (2013) Structural Yang, I., Clarke, M., Sim, E. and Fong, K. (2012)
basis of signaling blockade by anti-IL-13 antibody Inhaled corticosteroids for stable chronic obstructive
lebrikizumab. J Mol Biol 425: 13301339. pulmonary disease. Cochrane Database Syst Rev 7:
CD002991.
Wagener, A., de Nijs, S., Lutter, R., Sousa, A.,
Weersink, E., Bel, E. etal. (2015) External validation Ying, S., Robinson, D., Meng, Q., Rottman, J.,
of blood eosinophils, FENO and serum periostin as Kennedy, R., Ringler, D. etal. (1997) Enhanced
surrogates for sputum eosinophils in asthma. Thorax expression of eotaxin and CCR3 mRNA and
70: 115120. protein in atopic asthma. Association with airway
hyperresponsiveness and predominant co-localization
Wardlaw, A. (1999) Molecular basis for selective of eotaxin mRNA to bronchial epithelial and
eosinophil trafficking in asthma: a multistep paradigm. endothelial cells. Eur J Immunol 27: 35073516.
J Allergy Clin Immunol 104: 917926.
Zietkowski, Z., Bodzenta-Lukaszyk, A., Tomasiak,
Wardlaw, A., Silverman, M., Siva, R., Pavord, I. and M., Skiepko, R. and Szmitkowski, M. (2006)
Green, R. (2005) Multi-dimensional phenotyping: Comparison of exhaled nitric oxide measurement with
towards a new taxonomy for airway disease. Clin Exp conventional tests in steroid-naive asthma patients.
Allergy 35: 12541262. J Investig Allergol Clin Immunol 16: 239246.
Wenzel, S., Ford, L., Pearlman, D., Spector, S., ZuWallack, R., Mahler, D., Reilly, D., Church, N.,
Sher, L., Skobieranda, F. etal. (2013) Dupilumab in Emmett, A., Rickard, K. etal. (2001) Salmeterol plus Visit SAGE journals online
http://taj.sagepub.com
persistent asthma with elevated eosinophil levels. theophylline combination therapy in the treatment of
N Engl J Med 368: 24552466. COPD. Chest 119: 16611670. SAGE journals

http://taj.sagepub.com 51

Das könnte Ihnen auch gefallen