Asthma 2: Targeting The Interleukin Pathway in The Treatment of Asthma
Asthma 2: Targeting The Interleukin Pathway in The Treatment of Asthma
Asthma 2: Targeting The Interleukin Pathway in The Treatment of Asthma
Asthma 2
Targeting the interleukin pathway in the treatment of asthma
Kian Fan Chung
Lancet 2015; 386: 1086–96 Asthma is a common heterogeneous disease with a complex pathophysiology. Current therapies based on inhaled
See Editorial page 1014 corticosteroids and longacting β2 agonists are effective in controlling asthma in most, but not all patients, with a few
This is the second in a Series of patients falling into the severe asthma category. Severe asthma is characterised by poor asthma control, recurrent
two papers about Asthma exacerbations, and chronic airflow obstruction despite adequate and, in many cases, high-dose treatments. There is
Experimental Studies, Airway strong evidence supporting the role for interleukins derived from T-helper-2 (Th2) cells and innate lymphoid cells,
Disease Section, National Heart such as interleukins 4, 5, and 13, as underlying the eosinophilic and allergic inflammatory processes in nearly half of
and Lung Institute, Imperial
College London, London, UK
these patients. An anti-IgE antibody, omalizumab, which binds to circulating IgE, a product of B cells from the
and National Institute for actions of interleukin 4 and interleukin 13, is used as treatment for severe allergic asthma. Studies examining cytokine
Health Research (NIHR), blockers such as anti-interleukin-5, anti-interleukin-4Rα, and anti-interleukin-13 monoclonal antibodies in patients
Respiratory Biomedical with severe asthma with recurrent exacerbations and high blood eosinophil counts despite use of inhaled
Research Unit, Royal Brompton
& Harefield NHS Trust and
corticosteroids have reported improved outcomes in terms of exacerbations, asthma control, and forced expiratory
Imperial College London, volume in 1 s. The US Food and Drug Administration’s recommendation to use an anti-interleukin-5 antibody for the
London, UK treatment of severe eosinophilic asthma suggests that there will be a therapeutic place for these anti-Th2 agents.
(Prof Kian Fan Chung DSc) Biomarkers should be used to identify the right patients for such targeted approaches. More guidance will be needed
Correspondence to: as to which patients should receive each of these classes of selective antibody-based treatments. Currently, there is no
Prof Kian Fan Chung, National
Heart and Lung Institute,
treatment that targets the cytokines driving asthma associated with non-eosinophilic inflammation and low Th2
Imperial College London, expression.
Dovehouse St,
London SW3 6LY, UK Introduction slow-release theophylline, and longacting anti-
[email protected]
Asthma is the 14th most important chronic disease in the cholinergics can be added. Some patients can be offered
world in terms of the prevalence, extent, and duration of daily treatment with oral corticosteroids, whereas
disability, affecting 334 million individuals of all ages.1 treatment with the monoclonal anti-IgE antibody
Although mortality from asthma has been falling, the omalizumab may be indicated in those with an allergic
disease still accounts for 90 and 170 deaths per million in background. 5–10% of patients need such high-level
female and male individuals, respectively. The economic treatments4 and have severe asthma.5,6
costs of asthma are made up of direct costs such as To understand the processes that increase asthma
hospital care and treatment, and indirect costs such as severity, research has continued into the pathophysiology
time lost from work. In Europe, the total cost per patient of asthma, and has focused on the interleukins or
has been estimated to range from €509 for controlled cytokines and their potential role in the disease.7 (In this
asthma to €2281 for uncontrolled asthma.2 Series paper, cytokines and interleukins are terms used
Current management of asthma consists of non- interchangeably to indicate a broad and loose category of
pharmacological approaches, such as allergen avoidance, small proteins that are important in activating cell
and pharmacological approaches.3 Daily inhaled signalling in the context of the inflammatory process of
corticosteroids combined with longacting β2 agonists are asthma.) This Series paper will focus on how these
the mainstay of asthma treatment. For patients who cytokines drive the inflammatory processes and
remain uncontrolled on this combination, other pathophysiological mechanisms of asthma, and the
controller drugs such as leukotriene-receptor antagonists, results of blocking the effects of cytokines on asthma
outcome measures in patients with severe asthma,
particularly with approaches that block anticytokine
Search strategy and selection criteria
antibodies. It will also highlight the diversity of the
Data for this Series paper were identified by searches in inflammatory process in severe asthma such that it
MEDLINE and PubMed and references from relevant articles becomes important to define the phenotype that will
and reviews in English, using the search terms “asthma” or respond best to each specifically targeted treatment.
“asthma treatments” with “interleukins” and “cytokines”,
refining the search to specific interleukins or cytokines of Airway inflammation, cytokines and
interest between Jan 1, 2005 and June 30, 2015. Search was interleukins, and molecular phenotyping
also made in the abstract publications from the European Asthma is a multicellular disease. It is a chronic
Respiratory Congress held in 2014, and the American Thoracic inflammatory disease of the airways characterised by
Meeting held in May, 2014, and Sept, 2015. infiltration of eosinophils, basophils, mast cells, and
CD4 T-helper cells into the airway submucosa. This
inflammatory process is associated with mucus allergic eosinophilic asthma. They produce
hypersecretion, airway wall remodelling, and airway interleukins 4, 5, and 13, which leads to an increase in
hyper-responsiveness, which may all contribute to IgE production and eosinophilic inflammation, whereas
chronic airway obstruction. One important initiating innate immune cytokines such as interleukins 1, 25, and
component is the airway epithelium, along with 33, and thymic stromal lymphopoietin (TSLP) released
dendritic cells and airway macrophages, responding to from airway epithelial cells can activate Th2 cells or
external environmental factors such as allergens, ILC2s, or both.9
pollutants, and infectious agents (eg, viruses). This The heterogeneity of asthma has long been recognised,
initial response leads to the transmission of signals to both in terms of clinical outcomes and response to
other cells such as T cells, B cells, and mast cells, with available treatments. Unsupervised clustering methods
activation of airway structural cells such as airway with use of clinical features and inflammatory biomarkers
smooth muscle cells, mucus goblet cells, and fibroblasts8 have identified phenotypes described by asthma control
(figure 1). Cytokines are important in the promotion of and severity, age of onset of disease, obesity, and sputum
chronic airway inflammation and remodelling9,10 by their eosinophil count.13,14 This phenotypic diversity can be
actions on inflammatory and structural cells and by accounted for by different inflammatory mechanisms
affecting and promoting cell–cell interactions.11,12 underpinned by different molecular pathways.15 Because
T-helper-2 (Th2) cells and type 2 innate lymphoid cells Th2 cells have been recognised as an important part of the
(ILC2s) are now recognised as important cells underlying mechanisms underlying asthma, a Th2-high asthma has
Macrophage Epithelium
Interleukin 12
TNFα B7.2 CD28
Interleukin 4Rα Interleukin 4Rα Interleukin 4Rα
Interleukin 1β
Dendritic MHCII TCR Th0
cells
nity
mu
e im mal
In nat chy
m e sen n Interleukin 6
al- rmati o
heli TGFβ
Epit transfo TSLP, interleukin 33, interleukin 25
Severe asthma: poor asthma control, recurrent exacerbations, chronic airflow obstruction, corticosteroid insensitivity
Figure 1: Pathophysiological mechanisms underlying severe asthma and targets for anti-interleukin therapy
Interactions between environmental factors and innate and adaptive immune and inflammatory responses are initiated at the level of the airway epithelium, in
particular alveolar macrophages and dendritic cells, with the release of TSLP, interleukin 33, and interleukin 25 from the airway epithelium. Generation of Th2 cells
and ILC2s leads to the elaboration of interleukins 4, 5, and 13, which are important for the generation of an allergic eosinophilic inflammation with eosinophil
recruitment and activation, and the production of IgE. Activation of Th1 and Th17 cells can also be a feature of asthma, possibly contributing to neutrophilic
inflammation. Airway wall remodelling and repair driven by epithelial-mesenchymal transformation and the effects of interleukins on airway structural cells such as
airway smooth muscle cells and epithelial cells contribute to chronic airflow obstruction and bronchial hyper-responsiveness. Targets for interleukin intervention
against interleukins 4, 5, and 13 have included GATA3 in Th2 and ILC2 cells and antibody-directed inhibition against each individual interleukin or against relevant
receptors such as interleukin 4Rα or interleukin 5Rα. ILC2=type 2 innate lymphoid cells. TCR=T-cell receptor. Th1=T-helper-1 cell; Th2=T-helper-2 cell.
Th17=T-helper-17 cell. TFGβ=transforming growth factor β. TNFα=tumour necrosis factor α. TSLP=thymic stromal lymphopoietin.
GM-CSF=granulocyte-macrophage colony-stimulating factor.
been defined by use of genes expressed in airway epithelial of Th2 cells, synthesis of innate cytokines, such as TSLP,
brushings, consisting of a set of interleukin-13-inducible interleukin 25, and interleukin 33 from the epithelium,28
genes comprising POSTN, CLCA1, and SERPINB2.16 Only can be induced by external stimuli such as
half of a group of patients with mild-to-moderate asthma lipopolysaccharide, pollutants, and viruses.29 Additionally,
had a Th2-high asthma phenotype that was characterised these cytokines activate dendritic cells resulting in further
by a greater degree of bronchial hyper-responsiveness, differentiation and clonal expansion of Th2 cells with
higher serum IgE concentrations, and increased blood further activation of Th2 cells and tissue eosinophilia.
and airway eosinophilia.17 This finding has led to the Effector Th2 cells release interleukins 4, 5, and 13,
dichotomisation of asthma into a Th2-high and a Th2-low which are expressed in the bronchial submucosa of
endotype.18 This categorisation has implications for testing patients with asthma. An alternative source of
new compounds that target specific molecular pathways interleukin 5 and interleukin 13 is ILC2s, which express
in the clinic, particularly in identifying patients most likely no conventional lymphocyte or dendritic-cell phenotypic
to respond to these treatments.19 In the first clinical trial of markers. ILC2s require GATA3 transcription factor for
the anti-interleukin-5 antibody mepolizumab, which was their development and function; they are also stimulated
used to block the effects of the Th2-derived cytokine by TSLP and interleukins 25 and 33 to produce
interleukin 5, the drug proved ineffective in patients with interleukins 5 and 13.23 High concentrations of ILC2s
asthma recruited solely according to severity;20 however, in have been seen in eosinophilic nasal polyps, as well as in
a subsequent study in patients with severe asthma with a peripheral blood of patients with asthma.26,27
history of frequent exacerbations and eosinophilia in Although IgE is not considered an interleukin, its role
sputum or blood, mepolizumab treatment resulted in an in allergic inflammation is crucial and its production
important reduction in exacerbations compared with from B cells is a result of the effect of interleukin 4 and
placebo.21 interleukin 13 in causing a class switch in immunoglobulin
Biomarkers that are accessible in the clinic can be used synthesis towards IgE. After IgE binds to high-affinity
to stratify patients, especially for identification of those receptors on mast cells and basophils, cross-linking of
who will respond to blocking antibodies directed towards IgE receptors leads to subsequent activation of these cells
interleukins 4, 5, and 13. Eosinophil counts in blood are to release preformed mediators and cause synthesis of
positively correlated to eosinophil counts in sputum; cysteinyl leukotrienes and pro-inflammatory cytokines.
raised blood eosinophil counts of more than 220 cells per Thus, IgE-activated mast cells secrete interleukins, such
μL are present in 53% of patients with severe asthma and as interleukins 3, 4, and 5, and granulocyte-macrophage
sputum eosinophilia of more than 2% is present in 55% colony-stimulating factor, which recruit or activate
of patients with severe asthma.22 In patients with mild-to- eosinophils.
severe asthma, blood eosinophil count was also reported Omalizumab is a humanised antibody that binds to the
to have the highest accuracy in identifying sputum Cε3 domain of the free IgE heavy chain. When it binds to
eosinophilia in asthma, compared to nitric oxide free circulating IgE, there is a reduction in IgE available for
concentration in exhaled breath, or serum periostin.23 A binding to high-affinity IgE receptors (FcεR1) and also a
sputum eosinophilia of more than 2% was totally specific reduction in high-affinity FcεR1 receptors present on mast
but had a poor sensitivity for airway Th2 inflammation.24 cells and basophils.30 These events lead to an interruption
These findings suggest that blood eosinophil counts of the allergic cascade and prevention of mast-cell
alone might be useful in selecting individuals with Th2- degranulation with a reduction in eosinophilic inflam-
high asthma, and this approach has been used to indicate mation.30 Furthermore, omalizumab decreases early and
a population of patients with severe asthma who respond late asthmatic responses, sputum and tissue eosinophil
to interleukin-5 blocking agents.25 Another biomarker, counts, and submucosal cells positive for IgE and FcεR1.31–33
serum periostin, has been reported to be a better Omalizumab improved asthma-related quality-of-life
predictor of airway eosinophilic inflammation in patients measures and reduced exacerbation rates, with small
with severe asthma26 and has successfully identified improvements in FEV1 in patients with moderate-to-severe
patients with moderate-to-severe asthma who show an asthma.4,34 In severe persistent allergic asthma, omalizumab
improvement in forced expiratory volume in 1 s (FEV1) as an add-on therapy decreased clinically significant asthma
after treatment with an anti-interleukin-13 antibody.27 exacerbations by 26%, compared with placebo.35 Biomarkers
of therapeutic efficacy for omalizumab in reducing
Anti-interleukin approaches in asthma exacerbations are high concentrations of nitric oxide in
Targeting the Th2 pathway exhaled breath and high blood eosinophil counts.36 A trial
Activation of the Th2 pathway is initiated from a complex of omalizumab in both adults and children with severe
interaction between the innate and adaptive immune allergic asthma is recommended.4
responses, starting with the differentiation of Th2 cells More potent anti-IgE antibodies are in development.
from uncommitted T cells, which requires the action of QGE031 (Novartis, Basel, Switzerland) is a humanised
co-stimulatory molecules and cytokines expressed by anti-IgE antibody with a 50-fold higher affinity for IgE
dendritic cells and inflammatory cells. For the activation than omalizumab. QGE031 reduced circulating
count of 300 cells per μL or more, although not when single nebulised dose was shown to be effective in
given as a dose of 200 mg every 4 weeks.46 Dupilumab also moderate asthma and was associated with improvement in
reduced annualised severe exacerbation rates irrespective FEV1, lower use of β2 agonists, and reduced concentrations
of baseline eosinophil counts, apart from when given as a of exhaled nitric oxide compared with placebo.61 In a dosing
dose of 300 mg every 4 weeks. These findings might raise study over 12 weeks, there was significant effect in
questions about the role of blood eosinophils as a preventing reduction in FEV1 but not on exacerbation rates
predictive biomarker of response to this treatment. following reduction of maintenance doses of inhaled
corticosteroids, in moderate persistent asthma.62 However,
Anti-interleukin-4 antibody no benefits were reported in subsequent clinical trials.
Inhaled recombinant human soluble interleukin-4 There has been no further development of this soluble
receptor has been used to antagonise interleukin 4. A interleukin-4 receptor in asthma.
ACQ=Asthma Control Questionnaire. AQLQ=Asthma Quality of Life Questionnaire. DPP-4=dipeptidyl peptidase 4. FENO=exhaled nitric oxide concentration. FEV1=forced expiratory volume in 1 s. ICS=inhaled
corticosteroid; LABA=long acting β2 agonist. OCS=oral corticosteroid. ppb=parts per billion.
AMG 157 (Amgen, Thousand Oaks, CA, USA), a Brodalumab (Amgen, Thousand Oaks, CA, USA), a
human anti-TSLP monoclonal antibody, attenuated human anti-interleukin-17RA monoclonal antibody, had
allergen-induced early and late asthmatic responses in no effect on asthma control scores, symptom-free days,
mild allergic asthma. It also reduced blood and sputum and FEV1 in patients with inadequately controlled
eosinophil counts before and after allergen challenge, moderate-to-severe asthma who were receiving inhaled
and fractional exhaled nitric oxide concentrations.72 corticosteroid therapy. Furthermore, analysis of nine
prespecified patient subgroups showed a high-
Antagonists of CRTh2 reversibility subgroup with a post-bronchodilator FEV1
Prostaglandin D2, released during allergen-induced improvement of 20% or more who showed a significant
reactions in the airways, activates a G-protein-coupled improvement in ACQ score at the 210 mg but not at the
receptor, chemoattractant receptor-homologous molecule 280 mg dose.82 A follow-up phase 2b study focusing on
expressed on Th2 cells (CRTh2, also known as DP2 this phenotype was stopped because of a lack of reported
receptor), present on Th2 cells, ILC2s, and eosinophils, efficacy in an interim analysis. There has been no further
and induces the chemotaxis of these cells.73 Production of development of this antibody in asthma.
Th2-type cytokines by ILC2s is also stimulated by CXCL8 is a chemokine (chemoattractant cytokine)
prostaglandin D2 via activation of its CRTh2 receptor,24 involved in the chemoattraction and activation of
and also by leukotriene D4 (LTD4).25 neutrophils through the CXCR2 receptor.
In patients with moderate persistent asthma who were SCH527123 (Merck, Whitehouse Station, NJ, USA), a
not taking inhaled corticosteroids, the CRTh2 antagonist CXCR2 antagonist, inhibited ozone-induced sputum
OC000459 (Oxagen, Abingdon, UK) improved FEV1, neutrophilia in patients without asthma.83 It also reduced
asthma-related quality of life scores, and night-time sputum neutrophilia in adults with severe asthma, with a
symptoms, without affecting sputum eosinophil counts, modest reduction in mild exacerbations, but did not
compared with placebo.74 The CRTh2 antagonist AMG improve asthma control.84
853 (Amgen, Thousand Oaks, CA, USA), used as an add- Tumour necrosis factor α (TNFα) is a pro-inflammatory
on to inhaled corticosteroid therapy, did not improve Th1 cytokine that induces airway inflammation and hyper-
asthma symptoms or lung function in patients with responsiveness, mucus hypersecretion, and activation of
uncontrolled moderate-to-severe disease.75 Another macrophages. In two small, placebo-controlled, double-
CRTh2 antagonist, BI671800 (Boehringer Ingelheim, blind, crossover studies of patients with corticosteroid-
Ingelheim am Rhein, Germany), caused a small refractory asthma, treatment with etanercept (Pfizer,
improvement in FEV1 in symptomatic patients who had Collegeville, PA, USA) (a soluble recombinant dimer
not previously used controller treatment and in patients protein consisting of two human TNFα receptors fused
on inhaled corticosteroids.76 In a phase 2a study in patients with the Fc domain of human IgG1) resulted in clinical
with mild-to-severe asthma with sputum eosinophil improvements in PC20 (the concentration of metha-
counts of more than 2%, treatment with another CRTh2 choline required to provoke a 20% decrease in FEV1),
antagonist, QAW039 (Novartis, Basel, Switzerland), quality of life scores, and lung function in one study,85 and
resulted in a 3·5-fold reduction in sputum eosinophil improvements in ACQ scores with reductions in the use of
count compared with placebo, accompanied by a 2·3-fold nebulised β2 agonists in the other.86 However, in a
reduction in eosinophil count in bronchial biopsies.77 subsequent larger study in adults with uncontrolled severe
QAW039 improved AQLQ score, but not ACQ-7 score, persistent asthma, the anti-TNFα antibody golimumab
compared with placebo. (Centocor, Malvern, PA, USA) had no overall beneficial
effects. A post-hoc analysis suggested that patients
Targeting non-Th2 targets with substantial bronchodilator reversibility had fewer
Th17 cells are CD4 T cells that express interleukins 17A, exacerbations while on golimumab.87 Serious side-effects
17E, 17F, and 22, and are able to mediate neutrophil with golimumab included an increased frequency of
activation via the production of CXCL8 (interleukin 8).78,79 infections and malignancies compared with placebo,
Allergens and environmental stimuli such as cigarette which probably represents the importance of TNFα in the
smoke and diesel exhaust particles can trigger innate immune response to infections and cancer.
Th17-mediated airway inflammation in asthma.80
Overexpression of interleukin 17A and interleukin 17F Summary and conclusions
has been shown in lung tissue from patients with Blockade of the effects of interleukin 5 with a monoclonal
asthma, with expression levels correlating with asthma antibody directed towards interleukin 5 or its receptor,
severity, especially in patients with neutrophilic interleukin 5Rα, in patients with severe asthma with
corticosteroid-resistant disease.81 Interleukin 17A or eosinophilia in blood or sputum led to a reduction in
interleukin 17F, or both, stimulate airway structural cells, asthma exacerbation rates and an improvement in asthma
including bronchial epithelial cells and subepithelial control and FEV1 (table). Blocking the effects of
fibroblasts, to secrete neutrophil chemoattractants such interleukin 13 with anti-interleukin-13 antibody has led to
as CXCL8 and CXCL1. an improvement in FEV1, particularly in patients with a
high serum concentration of periostin, with a reduction in bacterial or viral infections, and linked to corticosteroid
exacerbations with lebrikizumab, but not with insensitivity, and also to airway wall remodelling.
tralokinumab. Blocking interleukin 4Rα with dupilumab, Additionally, there might be interactions between Th2
which neutralises the effects of interleukin 4 and and non-Th2 mechanisms.
interleukin 13, reduced exacerbations irrespective of Targeting the interleukin pathway has led to the
baseline eosinophil count, improved FEV1 in those with formulation of antibody therapies against interleukins 4,
high blood eosinophil count, and reduced serum IgE 5, and 13, which are important cytokines underlying the
concentrations. Interleukin 5, therefore, has a most eosinophilic inflammation in asthma. These treatments
important pathogenic role in exacerbations through its will hopefully become available for patients with severe
role in activating eosinophils, and the modest improvement asthma in the coming decade and will represent an
in airflow obstruction after antibody treatment perhaps important milestone in the management of severe
results from disruption of the interaction between asthma.
eosinophils and airway smooth muscle cells. Alternatively, Contributors
the improvement in FEV1 with anti-interleukin-13 antibody I am sole author and contributor.
might result from direct inhibition of airway smooth Declaration of interests
muscle contractility and bronchial hyper-responsiveness. I have received honoraria for participating in advisory board meetings
The reduction in exacerbations noted with anti-interleukin- regarding treatments for asthma and chronic obstructive pulmonary
disease for GlaxoSmithKline, AstraZeneca, Novartis, and Johnson &
4Rα antibody might result from inhibition of both Johnson, and have received research grant funding through my
interleukin 4 and interleukin 13, which could implicate the institution from Pfizer, GSK, and Merck. I have also been remunerated
biomarkers that were inhibited—namely, IgE and the for speaking engagements by AstraZeneca, Merck, and Novartis.
chemokines CCL17 and CCL26. Acknowledgments
With the recent recommendation by the US Food and Work carried out in my laboratory is funded by the UK Medical Research
Drug Administration for use of mepolizumab as an add-on Council, National Environmental Research Council UK, the US National
Institute of Environmental Health Sciences, and the NIHR Respiratory
maintenance treatment in patients aged 18 years or older Biomedical Research Unit, Royal Brompton & Harefield NHS Trust and
with severe eosinophilic asthma, other antibody-based Imperial College London, UK.
therapies will also be approved for severe asthma. References
Inevitably, the question will be asked as to what are the 1 Global Asthma Network. Global asthma report 2014. Global burden
most appropriate antibody-based treatments (including of disease due to asthma. http://www.globalasthmareport.org/
burden/burden.php (accessed July 29, 2015).
anti-IgE therapy) for patients with severe uncontrolled 2 Accordini S, Corsico AG, Braggion M, et al. The cost of persistent
asthma associated with eosinophilia. One might assume asthma in Europe: an international population-based study in
that patients with recurrent exacerbations would benefit adults. Int Arch Allergy Immunol 2013; 160: 93–101.
3 Global Initiative for Asthma. Global strategy for asthma
most from an anti-interleukin-5 approach, whereas patients management and prevention. 2015. http://www.ginasthma.org/
with more chronic airflow obstruction without recurrent documents/4 (accessed July 29, 2015).
exacerbations would benefit from an anti-interleukin-13 4 Chung KF, Wenzel SE, Brozek JL, et al. International ERS/ATS
guidelines on definition, evaluation and treatment of severe
approach, and patients with both chronic airflow asthma. Eur Respir J 2014; 43: 343–73.
obstruction and recurrent exacerbations might benefit 5 Moore WC, Bleecker ER, Curran-Everett D, et al, and the National
from an anti-interleukin-5 or anti-interleukin-4Rα Heart, Lung, Blood Institute’s Severe Asthma Research Program.
treatment. Alternatively, one could resort to the current Characterization of the severe asthma phenotype by the National
Heart, Lung, and Blood Institute’s Severe Asthma Research
concept of biomarkers and the knowledge gained so far Program. J Allergy Clin Immunol 2007; 119: 405–13.
from the clinical trials of these blocking antibodies. Thus, 6 European Network for Understanding Mechanisms of Severe
for anti-IgE treatment, it would be necessary to demonstrate Asthma. The ENFUMOSA cross-sectional European multicentre
study of the clinical phenotype of chronic severe asthma.
raised total serum IgE and the presence of circulating Eur Respir J 2003; 22: 470–77.
specific IgEs, and a high concentration of serum periostin 7 Adcock IM, Caramori G, Chung KF. New targets for drug
perhaps an indication for use of anti-interleukin-13 development in asthma. Lancet 2008; 372: 1073–87.
8 Lambrecht BN, Hammad H. Allergens and the airway epithelium
antibody. Direct comparison of the effects of one antibody response: gateway to allergic sensitization. J Allergy Clin Immunol 2014;
treatment against another and a search for better 134: 499–507.
biomarkers or combinations of biomarkers that will predict 9 Holgate ST. Innate and adaptive immune responses in asthma.
Nat Med 2012; 18: 673–83.
response and efficacy may be needed.
10 Erle DJ, Sheppard D. The cell biology of asthma. J Cell Biol 2014;
There has been less success with targets outside the 205: 621–31.
interleukin-4, interleukin-5, and interleukin-13 cytokines. 11 Chung KF, Barnes PJ. Cytokines in asthma. Thorax 1999;
The importance of the non-Th2 or low-Th2 expressors 54: 825–57.
remains unclear and there could be diverse mechanisms 12 Schuijs MJ, Willart MA, Hammad H, et al. Cytokine targets in
airway inflammation. Curr Opin Pharmacol 2013; 13: 351–61.
at play. What sputum neutrophilia is an indicator of is 13 Moore WC, Meyers DA, Wenzel SE, et al, and the National Heart,
not entirely clear, and endotyping of non-Th2 Lung, and Blood Institute’s Severe Asthma Research Program.
mechanisms in severe asthma is necessary. Non-Th2 Identification of asthma phenotypes using cluster analysis in the
Severe Asthma Research Program. Am J Respir Crit Care Med 2010;
mechanisms might involve excessive activation of the 181: 315–23.
innate inflammatory response, such as that induced by
14 Haldar P, Pavord ID, Shaw DE, et al. Cluster analysis and 38 Gauvreau GM, Harris JM, Boulet LP, et al. Targeting membrane-
clinical asthma phenotypes. Am J Respir Crit Care Med 2008; expressed IgE B cell receptor with an antibody to the M1 prime
178: 218–24. epitope reduces IgE production. Sci Transl Med 2014; 6: 243ra85.
15 Chung KF, Adcock IM. How variability in clinical phenotypes 39 Maes T, Joos GF, Brusselle GG. Targeting interleukin-4 in asthma:
should guide research into disease mechanisms in asthma. lost in translation? Am J Respir Cell Mol Biol 2012; 47: 261–70.
Ann Am Thorac Soc 2013; 10 (suppl): S109–17. 40 Wills-Karp M, Luyimbazi J, Xu X, et al. Interleukin-13: central
16 Woodruff PG, Boushey HA, Dolganov GM, et al. Genome-wide mediator of allergic asthma. Science 1998; 282: 2258–61.
profiling identifies epithelial cell genes associated with asthma and 41 Wenzel S, Wilbraham D, Fuller R, et al. Effect of an interleukin-4
with treatment response to corticosteroids. variant on late phase asthmatic response to allergen challenge in
Proc Natl Acad Sci USA 2007; 104: 15858–63. asthmatic patients: results of two phase 2a studies. Lancet 2007;
17 Woodruff PG, Modrek B, Choy DF, et al. T-helper type 2-driven 370: 1422–31.
inflammation defines major subphenotypes of asthma. 42 Slager RE, Otulana BA, Hawkins GA, et al. IL-4 receptor
Am J Respir Crit Care Med 2009; 180: 388–95. polymorphisms predict reduction in asthma exacerbations during
18 Wenzel SE. Asthma phenotypes: the evolution from clinical to response to an anti-IL-4 receptor alpha antagonist.
molecular approaches. Nat Med 2012; 18: 716–25. J Allergy Clin Immunol 2012; 130: 516–22.e4.
19 Chung KF. Asthma phenotyping: a necessity for improved 43 Wenzel SEIP, Otulana BA, Bleecker ER, et al. Inhaled pitrakinra,
therapeutic precision and new targeted therapies. J Intern Med 2015; an IL-4/IL-13 antagonist reduced exacerbations in patients with
published online June 15. DOI:10.1111/joim.12382. eosinophilic asthma. ERS annual congress 2010; Barcelona;
20 Flood-Page P, Swenson C, Faiferman I, et al, and the International Sept 18–22, 2010. Abstract 3980.
Mepolizumab Study Group. A study to evaluate safety and efficacy 44 Corren J, Busse W, Meltzer EO, et al. A randomized, controlled,
of mepolizumab in patients with moderate persistent asthma. phase 2 study of AMG 317, an IL-4Ralpha antagonist, in patients
Am J Respir Crit Care Med 2007; 176: 1062–71. with asthma. Am J Respir Crit Care Med 2010; 181: 788–96.
21 Pavord ID, Korn S, Howarth P, et al. Mepolizumab for severe 45 Wenzel S, Ford L, Pearlman D, et al. Dupilumab in persistent
eosinophilic asthma (DREAM): a multicentre, double-blind, asthma with elevated eosinophil levels. N Engl J Med 2013;
placebo-controlled trial. Lancet 2012; 380: 651–59. 368: 2455–66.
22 Schleich F, Brusselle G, Louis R, et al. Heterogeneity of phenotypes 46 Wenzel SE, Wang L, Pirozzi G, et al. Dupilumab improves lung
in severe asthmatics. The Belgian Severe Asthma Registry (BSAR). function and reduces severe exacerbations in uncontrolled asthmatics
Respir Med 2014; 108: 1723–32. with baseline eosinophil levels above and below 300 cells/μl. 2015
23 Wagener AH, de Nijs SB, Lutter R, et al. External validation of blood Meeting of the American Thoracic Society; Denver, CO, USA;
eosinophils, FE(NO) and serum periostin as surrogates for sputum May 15–20, 2015. Abstract A6362.
eosinophils in asthma. Thorax 2015; 70: 115–20. 47 Scheerens H, Arron JR, Zheng Y, et al. The effects of lebrikizumab
24 Peters MC, Mekonnen ZK, Yuan S, et al. Measures of gene in patients with mild asthma following whole lung allergen
expression in sputum cells can identify TH2-high and TH2-low challenge. Clin Exp Allergy 2014; 44: 38–46.
subtypes of asthma. J Allergy Clin Immunol 2014; 133: 388–94, e5. 48 Hanania NA, Noonan M, Corren J, et al. Lebrikizumab in
25 Katz LE, Gleich GJ, Hartley BF, et al. Blood eosinophil count is a moderate-to-severe asthma: pooled data from two randomised
useful biomarker to identify patients with severe eosinophilic placebo-controlled studies. Thorax 2015; 70: 748–56.
asthma. Ann Am Thorac Soc 2014; 11: 531–36. 49 Piper E, Brightling C, Niven R, et al. A phase II placebo-controlled
26 Jia G, Erickson RW, Choy DF, et al. Periostin is a systemic study of tralokinumab in moderate-to-severe asthma.
biomarker of eosinophilic airway inflammation in asthmatic Eur Respir J 2013; 41: 330–38.
patients. J Allergy Clin Immunol 2012; 130: 647–54, e10. 50 Brightling CE, Chanez P, Leigh R, et al. Efficacy and safety of
27 Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab tralokinumab in patients with severe uncontrolled asthma:
treatment in adults with asthma. N Engl J Med 2011; 365: 1088–98. a randomised, double-blind, placebo-controlled, phase 2b trial.
28 Lloyd CM, Saglani S. Epithelial cytokines and pulmonary allergic Lancet Respir Med 2015; published online July 28. http://dx.doi.
inflammation. Curr Opin Immunol 2015; 34: 52–58. org/10.1016/S2213-2600(15)00197-6.
29 Licona-Limón P, Kim LK, Palm NW, et al. TH2, allergy and group 2 51 De Boever EH, Ashman C, Cahn AP, et al. Efficacy and safety of an
innate lymphoid cells. Nat Immunol 2013; 14: 536–42. anti-IL-13 mAb in patients with severe asthma: a randomized trial.
J Allergy Clin Immunol 2014; 133: 989–96.
30 Djukanović R, Wilson SJ, Kraft M, et al. Effects of treatment with
anti-immunoglobulin E antibody omalizumab on airway inflammation 52 Leckie MJ, ten Brinke A, Khan J, et al. Effects of an interleukin-5
in allergic asthma. Am J Respir Crit Care Med 2004; 170: 583–93. blocking monoclonal antibody on eosinophils, airway
hyper-responsiveness, and the late asthmatic response. Lancet 2000;
31 Fahy JV, Fleming HE, Wong HH, et al. The effect of an anti-IgE
356: 2144–48.
monoclonal antibody on the early- and late-phase responses to
allergen inhalation in asthmatic subjects. 53 Haldar P, Brightling CE, Hargadon B, et al. Mepolizumab and
Am J Respir Crit Care Med 1997; 155: 1828–34. exacerbations of refractory eosinophilic asthma. N Engl J Med 2009;
360: 973–84.
32 van Rensen EL, Evertse CE, van Schadewijk WA, et al. Eosinophils
in bronchial mucosa of asthmatics after allergen challenge: effect of 54 Nair P, Pizzichini MM, Kjarsgaard M, et al. Mepolizumab for
anti-IgE treatment. Allergy 2009; 64: 72–80. prednisone-dependent asthma with sputum eosinophilia.
N Engl J Med 2009; 360: 985–93.
33 Djukanovic R, Wilson SJ, Kraft M, et al. Effect of treatment with
anti-IgE antibody (omalizumab) on airway inflammation in mild 55 Ortega HG, Liu MC, Pavord ID, et al, and the MENSA
atopic asthma. American Thoracic Society 99th international Investigators. Mepolizumab treatment in patients with severe
conference; Seattle, WA, USA; May 16–21, 2003. Abstract 703. eosinophilic asthma. N Engl J Med 2014; 371: 1198–207.
34 Normansell R, Walker S, Milan SJ, et al. Omalizumab for asthma in 56 Bel EH, Wenzel SE, Thompson PJ, et al, and the SIRIUS
adults and children. Cochrane Database Syst Rev 2014; 1: CD003559. Investigators. Oral glucocorticoid-sparing effect of mepolizumab in
eosinophilic asthma. N Engl J Med 2014; 371: 1189–97.
35 Humbert M, Beasley R, Ayres J, et al. Benefits of omalizumab as
add-on therapy in patients with severe persistent asthma who are 57 Castro M, Mathur S, Hargreave F, et al, and the Res-5-0010 Study
inadequately controlled despite best available therapy (GINA 2002 Group. Reslizumab for poorly controlled, eosinophilic asthma:
step 4 treatment): INNOVATE. Allergy 2005; 60: 309–16. a randomized, placebo-controlled study.
Am J Respir Crit Care Med 2011; 184: 1125–32.
36 Hanania NA, Wenzel S, Rosén K, et al. Exploring the effects of
omalizumab in allergic asthma: an analysis of biomarkers in the 58 Castro M, Zangrilli J, Wechsler ME, et al. Reslizumab for
EXTRA study. Am J Respir Crit Care Med 2013; 187: 804–11. inadequately controlled asthma with elevated blood eosinophil
counts: results from two multicentre, parallel, double-blind,
37 Arm JP, Bottoli I, Skerjanec A, et al. Pharmacokinetics,
randomised, placebo-controlled, phase 3 trials.
pharmacodynamics and safety of QGE031 (ligelizumab), a novel
Lancet Respir Med 2015; 3: 355–66.
high-affinity anti-IgE antibody, in atopic subjects.
Clin Exp Allergy 2014; 44: 1371–85.
59 Castro M, Wenzel SE, Bleecker ER, et al. Benralizumab, an 74 Barnes N, Pavord I, Chuchalin A, et al. A randomized, double-blind,
anti-interleukin 5 receptor α monoclonal antibody, versus placebo placebo-controlled study of the CRTH2 antagonist OC000459 in
for uncontrolled eosinophilic asthma: a phase 2b randomised moderate persistent asthma. Clin Exp Allergy 2012; 42: 38–48.
dose-ranging study. Lancet Respir Med 2014; 2: 879–90. 75 Busse WW, Wenzel SE, Meltzer EO, et al. Safety and efficacy of the
60 Nowak RM, Parker JM, Silverman RA, et al. A randomized trial of prostaglandin D2 receptor antagonist AMG 853 in asthmatic
benralizumab, an antiinterleukin 5 receptor α monoclonal antibody, patients. J Allergy Clin Immunol 2013; 131: 339–45.
after acute asthma. Am J Emerg Med 2015; 33: 14–20. 76 Hall IP, Fowler AV, Gupta A, et al. Efficacy of BI 671800, an oral
61 Borish LC, Nelson HS, Lanz MJ, et al. Interleukin-4 receptor in CRTH2 antagonist, in poorly controlled asthma as sole controller
moderate atopic asthma. A phase I/II randomized, placebo- and in the presence of inhaled corticosteroid treatment.
controlled trial. Am J Respir Crit Care Med 1999; 160: 1816–23. Pulm Pharmacol Ther 2015; 32: 37–44.
62 Borish LC, Nelson HS, Corren J, et al, and the IL-4R Asthma Study 77 Berair RGS, Singapuri A, Hartley R, et al. Effect of QAW039, an oral
Group. Efficacy of soluble IL-4 receptor for the treatment of adults prostaglandin D2 receptor (DP2/CrTh2) antagonist upon sputum
with asthma. J Allergy Clin Immunol 2001; 107: 963–70. and bronchial eosinophilic inflammation and clinical outcomes in
63 Noonan M, Korenblat P, Mosesova S, et al. Dose-ranging study of treatment-resistant asthma: a phase 2a randomised placebo-
lebrikizumab in asthmatic patients not receiving inhaled steroids. controlled trial. 2015 Meeting of the American Thoracic Society;
J Allergy Clin Immunol 2013; 132: 567–74, e12. Denver, CO, USA; May 15–20, 2015. Abstract A6361.
64 Nair P. Anti-interleukin-5 monoclonal antibody to treat severe 78 Roussel L, Houle F, Chan C, et al. IL-17 promotes p38 MAPK-
eosinophilic asthma. N Engl J Med 2014; 371: 1249–51. dependent endothelial activation enhancing neutrophil recruitment
65 Kolbeck R, Kozhich A, Koike M, et al. MEDI-563, a humanized to sites of inflammation. J Immunol 2010; 184: 4531–37.
anti-IL-5 receptor alpha mAb with enhanced antibody-dependent 79 Halwani R, Al-Muhsen S, Hamid Q. T helper 17 cells in airway
cell-mediated cytotoxicity function. J Allergy Clin Immunol 2010; diseases: from laboratory bench to bedside. Chest 2013; 143: 494–501.
125: 1344–53, e2. 80 Vroman H, van den Blink B, Kool M. Mode of dendritic cell
66 Kouzaki H, O’Grady SM, Lawrence CB, et al. Proteases induce activation: the decisive hand in Th2/Th17 cell differentiation.
production of thymic stromal lymphopoietin by airway epithelial Implications in asthma severity? Immunobiology 2015; 220: 254–61.
cells through protease-activated receptor-2. J Immunol 2009; 81 Al-Ramli W, Préfontaine D, Chouiali F, et al. T(H)17-associated
183: 1427–34. cytokines (IL-17A and IL-17F) in severe asthma.
67 Bleck B, Grunig G, Chiu A, et al. MicroRNA-375 regulation of J Allergy Clin Immunol 2009; 123: 1185–87.
thymic stromal lymphopoietin by diesel exhaust particles and 82 Busse WW, Holgate S, Kerwin E, et al. Randomized, double-blind,
ambient particulate matter in human bronchial epithelial cells. placebo-controlled study of brodalumab, a human anti-IL-17
J Immunol 2013; 190: 3757–63. receptor monoclonal antibody, in moderate to severe asthma.
68 Liu YJ, Soumelis V, Watanabe N, et al. TSLP: an epithelial cell Am J Respir Crit Care Med 2013; 188: 1294–302.
cytokine that regulates T cell differentiation by conditioning 83 Holz O, Khalilieh S, Ludwig-Sengpiel A, et al. SCH527123, a novel
dendritic cell maturation. Annu Rev Immunol 2007; 25: 193–219. CXCR2 antagonist, inhibits ozone-induced neutrophilia in healthy
69 Préfontaine D, Nadigel J, Chouiali F, et al. Increased IL-33 subjects. Eur Respir J 2010; 35: 564–70.
expression by epithelial cells in bronchial asthma. 84 Nair P, Gaga M, Zervas E, et al, and the Study Investigators. Safety
J Allergy Clin Immunol 2010; 125: 752–54. and efficacy of a CXCR2 antagonist in patients with severe asthma
70 Ying S, O’Connor B, Ratoff J, et al. Expression and cellular and sputum neutrophils: a randomized, placebo-controlled clinical
provenance of thymic stromal lymphopoietin and chemokines in trial. Clin Exp Allergy 2012; 42: 1097–103.
patients with severe asthma and chronic obstructive pulmonary 85 Berry MA, Hargadon B, Shelley M, et al. Evidence of a role of tumor
disease. J Immunol 2008; 181: 2790–98. necrosis factor alpha in refractory asthma. N Engl J Med 2006;
71 Wu J, Liu F, Zhao J, et al. Thymic stromal lymphopoietin promotes 354: 697–708.
asthmatic airway remodelling in human lung fibroblast cells 86 Morjaria JB, Chauhan AJ, Babu KS, et al. The role of a soluble
through STAT3 signalling pathway. Cell Biochem Funct 2013; TNFalpha receptor fusion protein (etanercept) in corticosteroid
31: 496–503. refractory asthma: a double blind, randomised, placebo controlled
72 Gauvreau GM, O’Byrne PM, Boulet LP, et al. Effects of an trial. Thorax 2008; 63: 584–91.
anti-TSLP antibody on allergen-induced asthmatic responses. 87 Wenzel SE, Barnes PJ, Bleecker ER, et al, and the T03 Asthma
N Engl J Med 2014; 370: 2102–10. Investigators. A randomized, double-blind, placebo-controlled study
73 Pettipher R, Hansel TT, Armer R. Antagonism of the prostaglandin of tumor necrosis factor-alpha blockade in severe persistent asthma.
D2 receptors DP1 and CRTH2 as an approach to treat allergic Am J Respir Crit Care Med 2009; 179: 549–58.
diseases. Nat Rev Drug Discov 2007; 6: 313–25.