You are on page 1of 10

COPD: Journal of Chronic Obstructive Pulmonary Disease

ISSN: 1541-2555 (Print) 1541-2563 (Online) Journal homepage: http://www.tandfonline.com/loi/icop20

Biologic Drugs: A New Target Therapy in COPD?

Ahmed Yousuf & Christopher E Brightling

To cite this article: Ahmed Yousuf & Christopher E Brightling (2018): Biologic Drugs: A New
Target Therapy in COPD?, COPD: Journal of Chronic Obstructive Pulmonary Disease, DOI:
10.1080/15412555.2018.1437897

To link to this article: https://doi.org/10.1080/15412555.2018.1437897

Published online: 23 Apr 2018.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=icop20
COPD: JOURNAL OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE
, VOL. , NO. , –
https://doi.org/./..

Biologic Drugs: A New Target Therapy in COPD?


Ahmed Yousuf and Christopher E Brightling
NIHR Leicester Biomedical Research Centre, Institute for Lung Health, Department of Infection, Immunity & Inflammation, University of Leicester,
Leicester, United Kingdom

ABSTRACT ARTICLE HISTORY


Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease associated with significant mor- Received  January 
bidity and mortality. Current diagnostic criteria based on the presence of fixed airflow obstruction and Accepted  February 
symptoms do not integrate the complex pathological changes occurring within the lung and they do not KEYWORDS
define different airway inflammatory patterns. The current management of COPD is based on ‘one size fits COPD; biologics; eosinophil;
all’ approach and does not take the importance of heterogeneity in COPD population into account. The anti-IL; benralizumab;
available treatments aim to alleviate symptoms and reduce exacerbation frequency but do not alter the mepolizumab; anti-TNFα;
course of the disease. Recent advances in molecular biology have furthered our understanding of inflam- anti-IL; anti-ILβ, IL, IL;
matory pathways in pathogenesis of COPD and have led to development of targeted therapies (biologics TSLP
and small molecules) based on predefined biomarkers. Herein we shall review the trials of biologics in COPD
and potential future drug developments in the field.

Introduction Pathophysiology
Chronic obstructive pulmonary disease (COPD) is defined by COPD is a heterogeneous disease, which is a consequence
airflow obstruction that is not fully reversible and that is usu- of complex host-environment interactions over time as sum-
ally progressive (1). Cigarette smoke is the main causative agent, marized in Figure 1. Smoking together with other pollutants,
although other exposures (e.g. air pollution, occupational and pathogens and in some cases allergens insult the lung promoting
biomass fuel) are increasingly being recognised as important airway inflammation and damage in a susceptible host (7,8).
(2–4). COPD is characterized by persistent symptoms punc- This chronic airway inflammation and remodeling leads to
tuated by episodes of worsening symptoms beyond day-to-day small airway obliteration and emphysema, which in turn cause
variability known as exacerbations. airflow obstruction and clinical expression of the disease.
COPD is common and causes significant morbidity and
mortality. There are approximately 400 million cases of COPD
Airway inflammation
worldwide (5) and it is predicted to become the 3rd lead-
ing cause of mortality worldwide by 2030 (4). In the United The presence of airway inflammation in both stable COPD
Kingdom it is responsible for approximately 30,000 deaths and exacerbation episodes is well recognised, with severity of
per year and for >£800 million per year in direct health care airflow obstruction correlating with airway inflammation (8).
costs (6). The inflammatory profile in COPD is typically associated with
COPD is a heterogeneous condition with respect to clinical increased CD8+ T-cells and neutrophils (9,10). In some cases
presentation, underlying pathophysiology, disease progres- the neutrophilic response is a consequence of chronic airway
sion and response to therapy (7). Current therapies alleviate infection particularly a predominance of proteobacteria such as
symptoms and reduce exacerbations, but have little or no effect Hemophilus Influenzae (11). In a subgroup of 15–40% COPD
upon disease progression and follow a broadly ‘one size fits all’ patients there is eosinophilic inflammation (12). Eosinophils
approach to the disease. Greater understanding of the inflam- have been found in sputum, Broncho-Alveolar Lavage (BAL)
matory mechanisms in COPD has led to advances in targeted fluid and tissue (12). An increase in the number of eosinophils
biologic therapies for specific cytokines or their receptors. The has been found during exacerbations (13). In asthma this is
success of such approaches is predicated on selection of a patient predominately due to allergic mediated type-2 immunity with
group in which the intervention will be both safe and effective a possible contribution independent from allergy mediated
and that the therapy will be affordable. We shall consider herein by type-2 innate lymphoid cells (14). In COPD the cause of
the heterogeneity of the underlying airway inflammation and eosinophilic inflammation is uncertain. The possible trig-
remodeling and the success and failures of biological therapy to gers and cytokine networks involved in neutrophilic versus
date in COPD. eosinophilic COPD are described in Figure 2.

CONTACT Professor Christopher E Brightling ceb@le.ac.uk Institute for Lung Health, University Hospitals of Leicester, Leicester, LE QP, UK.
Color versions of one or more of the figures in this article can be found online at www.tandfonline.com/icop.
©  Taylor & Francis Group, LLC
2 A. YOUSUF AND C. E. BRIGHTLING

COPD severity (29). These squamous cells express increased


Interleukin 1 beta (IL-1β) which induces a fibrotic response
in adjacent airway fibroblasts and is implicated in activation
of transforming growth factor (TGF)-β. TGF-β is a potent
fibrogenic factor that is increased in the small airway epithe-
lial cells in COPD. This activation of TGF-β correlates with
disease severity and small airway thickening in COPD (30,31).
The EGF and TGF-β released from small airways may be
involved in fibroblast activation and proliferation. Increased
bronchial deposition of extracellular matrix proteins includ-
ing collagens, fibronectin and laminin is observed with depo-
Figure . COPD is a heterogeneous complex disease as a consequence of complex sition increased on the epithelial basement membrane at sites
host-environment interactions across spatial scales within the host over time. of damage. The negative correlation between bronchial extra-
cellular matrix deposition and FEV1 supports the view that
bronchial deposition of extracellular matrix is related to airway
remodeling (32).
Airway remodeling
Airway remodeling refers to structural changes that occur in
Lung damage
small and large airways. Remodeling changes that occur in
COPD include disruption and loss of cilia, squamous metapla- In emphysema activated neutrophils release neutrophil elastase,
sia of the respiratory epithelium, goblet cell hyperplasia and a proteinase that can destroy elastin (33). In healthy individuals,
mucous gland enlargement, bronchiolar smooth muscle hyper- elastase is neutralized by alpha-1-antitrypsin, a proteinase
plasia and hypertrophy, airway wall fibrosis and inflammatory inhibitor that protects lung tissue from neutrophil elastase
cell infiltration (15,16). Small airways are the major site of airway damage. Alpha-1-antitrypsin deficiency leads to an imbalance
obstruction in COPD. The airways obstruction in COPD is due between proteinases and anti-proteinases, which in turn leads
to a combination of remodeling and accumulation of inflamma- to uncontrolled elastin destruction resulting in parenchymal
tory exudates within the airway lumen (17,18). There is some lung destruction (34).
evidence that a reduction in number and luminal area of distal Macrophages and neutrophils are found in greater numbers
airways is the cause of increased peripheral airway resistance in in COPD airways (35), and they release elastolytic proteinases
COPD (19). (e.g. matrix-metalloproteinases, cathepsins and collagenases)
Cigarette smoke is a key trigger for tissue damage and is also when activated, which lead to degradation of the extra-cellular
implicated in the abnormal lung repair processes seen in COPD matrix.
(20). Amongst the multiple effects of cigarette smoking, smok-
ing causes increased epithelial permeability (21) and can activate
Current pharmacological therapies in stable COPD
epidermal growth factor (EGF) receptors. Increased EGF and
EGF receptor (EGFR) expression is seen in bronchial epithelium The two main approaches for current drugs in management of
in COPD and has been implicated in the remodeling processes stable COPD are bronchodilators (e.g. β 2 agonists, antimus-
(22–25). Activation of EGFR can lead to mucin synthesis and carinics, and methylxanthines) and anti-inflammatories (e.g.
goblet cell hyperplasia, and EGF is also a stimulator of airway corticosteroids). Current recommendations base treatment
smooth muscle proliferation (26). It has been shown that there decisions on COPD control determined by symptoms scores
is a negative correlation between smooth muscle in the small air- e.g. Modified Medical Research Council (mMRC), COPD
ways and Forced Expiratory Volume in 1 second (FEV1) (27,28). Assessment Tool (CAT) and Clinical COPD Questionnaire
Epithelial changes observed in COPD include small air- (CCQ) and future risk of disease progression and exacerba-
way squamous metaplasia, which increases with increasing tions predicted by current lung function and exacerbation

Figure . Cytokine networks in COPD, in those with eosinophilic disease or non-eosinophilic predominately neutrophilic disease. Biologics that have shown promise and
are in phase  are shown in blue boxes, those that are in early phase trials in green boxes and those that have been unsuccessful are in orange boxes.
COPD: JOURNAL OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE 3

lung function, health status and walking distance (36–38).


Management directing corticosteroid therapy in stable disease
to minimize eosinophilic inflammation has led to reduced
hospitalization as observed also in asthma (39). Interestingly
the blood eosinophil count is consistently correlated with the
sputum eosinophil count and an increased blood eosinophil
count is associated with greater improvements to inhaled cor-
ticosteroids in stable disease and oral corticosteroids in acute
exacerbations (40,41). Thus, it is likely in the near future that
this measure will become part of standard-of-care in directing
corticosteroid therapy in COPD.

Biologics in COPD
Figure . Summary of drug treatment based on GOLD disease classification. (SABA With the neutrophil and pro-inflammatory cytokines implicated
= short acting beta agonist, LAMA = long acting muscarinic antagonist, LABA = in the pathogenies of COPD the first biologics in COPD have
long acting beta agonist, ICS = inhaled corticosteroid.)
targeted this element of the inflammatory response. The efficacy
has been disappointing and inhibiting these cytokines has led to
frequency (1). Phenotype-specific approaches to management adverse events which has diminished enthusiasm for biologics
of COPD are used in clinical practice as per Global Initiative for in COPD (42–44). In contrast, targeting eosinophilic inflamma-
Chronic Obstructive Lung Disease (GOLD) recommendations tion in those with evidence of this phenotype has demonstrated
as shown in Figure 3, but do not include measures of underlying efficacy without an increase in adverse events (45,46). These data
pathobiology. are more promising and have begun to pave a way forward for
As described above, eosinophilic inflammation is present in biologics in COPD. The Phase 2 and 3 studies of biologics in
a subset of COPD patients and an elevated sputum eosinophil COPD that have completed and reported their findings are as
count is associated with a greater response to inhaled and oral shown in Table 1 and how they map onto the cytokine networks
corticosteroids in stable disease in terms of improvement in is as shown in Figure 2.
Table . Completed and reported phase  and  trials of biologics in COPD (Anti-IL = anti-interleukin , Anti-TNF-α = anti- tumour necrosis factor alpha, Anti-IL = Anti-
interleukin , IL = Interleukin , CRQ = Chronic respiratory questionnaire, TDI = Transitional Dyspnoea Index, SGRQ-C = St. George’s Respiratory Questionnaire for COPD).

Drug, Dose & Duration Population Primary outcome Secondary outcome

Anti- Pro-inflammatory, neutrophilic, non-T2 inflammation


Anti-IL Mod-severe COPD (n = ) ↓ Severity of dyspnoea as ↔ Health status, lung function, -min
measured by TDI walk test, rescue use of albuterol
 mg loading dose,  mg/month for > years, MRC ࣙ
 months,  month follow-up ()
> pack year smoking history
Anti-TNFα (Etanercept) ࣙ years old (n = ) ↔ FEV over  days from ↔ -day treatment failure, dyspnoea,
exacerbation onset health status
 mg,  days () Enrolled at exacerbation onset
Anti-TNFα (Infliximab) Mild-mod COPD (n = ) ↔ Sputum inflammatory cells ↔ FEV , SGRQ
mg/kg,  weeks () Current smokers
Anti-TNFα (Infliximab) Mod-severe COPD (n = ) ↔ CRQ ↔ FEV , -min walk test, TDI
 mg/kg or  mg/kg,  weeks () > years, CRQ< ↑ Malignancy, pneumonia
Anti-IL (MEDI ) Mod-very severe COPD (n = ) ↔ Moderate-to-severe ↔ SGRQ-C
exacerbations
 mg every  weeks,  weeks () – years, ࣙ exacerbations in past year
Anti- eosinophilic, T2 inflammation
Benralizumab/ IL- Mod-severe COPD (n = ) ↔ Moderate-to-severe ↑ FEV in intervention group
exacerbations
 mg every  weeks ( doses) then every – years, ࣙ exacerbation in past year ↔ health status
 weeks ( doses),  week ()
Sputum eosinophils ࣙ% ↓ Blood and sputum eosinophils
Mepolizumab/ IL Mod-severe COPD (n = ) ↓ Sputum eosinophils ↓ Blood eosinophil
 mg/month,  months () Sputum eosinophils >% ↔ FEV , CAT, CRQ, exacerbations
ࣙ exacerbation in previous year
Mepolizumab/ IL- Mod-very severe COPD, (n = ) ↓ Exacerbations in pre-specified ↓ Time to first exacerbation
eosinophilic group
 mg or  mg every  weeks,  weeks > years, ࣙ moderate or ࣙ severe ↔ FEV , SGRQ, CAT
() exacerbation in previous year
Mepolizumab/ IL- Mod-very severe COPD, (n = ), ↓ Exacerbations ↓ Time to first exacerbation
> years, ࣙ moderate or ࣙ severe
exacerbation in previous year, blood
eosinophils >/µL
 mg or  mg every  weeks,  weeks ↔ FEV , SGRQ, CAT
()
4 A. YOUSUF AND C. E. BRIGHTLING

Anti- Pro-inflammatory, neutrophilic, non-T2 inflammation necrosis, apoptosis and oxidative stress. Biologics targeting
TNF-α have been very successful and indeed transformed treat-
Anti-IL and anti-CXCR
ment of several chronic inflammatory conditions including
The chemokine C-X-C Ligand 8 (CXCL8) (IL-8) is produced by
rheumatoid arthritis and inflammatory bowel disease (48).
a variety of cells, including monocytes, macrophages and neu-
In an observational study by Suissa and colleagues, etan-
trophils. It mainly attracts and activates neutrophils during an
ercept but not infliximab reduced COPD hospitalisation in a
inflammatory response (44). It binds to CXC chemokine recep-
group of patients who had rheumatoid arthritis and COPD (49).
tor 1 (CXCR1) and CXCR2 members of the G-protein coupled
The relative risk of COPD hospitalisation with the use of etan-
receptor family. CXCR2 is present on the surface of neutrophils
ercept was 0.47 and with infliximab 1.14. In this study, only 16
and is involved in their recruitment to the lung (44).
out of 1205 patient were on etanercept and 21 were on inflix-
Mahler and colleagues undertook a 3-month pilot study of
imab. Patients were identified from insurance claim data base,
an IL-8 monoclonal antibody in 109 subjects with COPD (47).
and therefore, it is not certain all the patients had lung function
Neutralization of IL-8 led to small but significant improve-
confirmed COPD. Notwithstanding this limitation, this pro-
ments in dyspnea measured using the transitional dyspnea index
vides some further support to study biologics against TNF-α in
(TDI). The study was not powered to detect differences in exac-
COPD.
erbation rate between the groups. This biologic was not pro-
Van der Vaart and colleagues undertook the first study of
gressed but gave encouragement to test inhibition of CXCR2
infliximab for 8 weeks in 22 subjects with COPD (50). There
with small molecule inhibitors. The major concern with this
was no increase in adverse events in those receiving infliximab
target is whether it might cause clinically important neutrope-
but also no benefits in lung function nor health status were
nia and subsequent predisposition of infection. Kirsten and
observed. This led to a larger study of infliximab in 234 sub-
colleagues undertook a 4-week study in 87 COPD subjects to
jects with COPD. In this study, there was no improvement in
investigate the safety and tolerability of a CXCR2 antagonist
health status, lung function, symptoms nor exacerbation fre-
(AZD5069) (44). The main adverse events reported were neu-
quency in those receiving infliximab versus placebo. Impor-
tropenia (<1× 109 /L) in intervention arm (3 patients in 50-
tantly there were more adverse events in those receiving inflix-
mg arm and 1 patient in 80-mg arm), and COPD exacerba-
imab with an increase in incidences of cancer (12 versus 3; 6
tions (1 in 50-mg arm and 1 in 80-mg arm). However, there was
lung cancers, 2 head and neck cancers, 1 breast cancer, 1 renal
no increase in infection in those receiving anti-CXCR2 versus
cell cancer, 1 Hodgkin’s lymphoma, 1 liver or pancreatic cancer
placebo. Rennard and colleagues undertook a 6-month dose-
versus 2 prostate cancer, 1 cervical cancer) and pneumonia (10
ranging proof-of-concept study of the CXCR2 antagonist MK-
versus 1). This difference in cancer risk during the study did not
7123 in 616 subjects with moderate-to-severe COPD (43). They
persist in the long-term follow-up of patients after the treatment
found a small significant improvement in FEV1 in those receiv-
cessation. However, the poor efficacy for anti-TNF-α in stable
ing the highest dose of anti-CXCR2 (50 mg daily) versus placebo.
disease and the increased cancer and infection risk has stopped
The benefit was observed in those that were current smokers
progress of this biologic as maintenance therapy.
with an improvement above placebo of 160 mL. There was also
Even though the poor safety of anti-TNF-α precludes its use
a reduction in the likelihood (hazards ratio 0.5) of exacerbation
in stable COPD, Aaron and colleagues undertook an acute study
in current smokers compared to placebo. Importantly, there was
of etanercept at the onset of an exacerbation versus prednisolone
no statistically significant difference in FEV1 or exacerbations
in 81 subjects with COPD. They found no increase in adverse
between ex-smokers in the intervention arm and placebo. The
events but neither did they find benefit in favor of etanercept.
study was terminated early at 12 months because not enough
No further studies of anti-TNF-α in COPD are ongoing.
patients continued with the study to the scheduled 18 months.
There was no difference in rate of infection between the two
groups at 6 months, however, in patients who continued with
Anti-interleukin  (anti- IL)
treatment beyond 6 months, the frequency of total infection and
IL-1 has been described as the master cytokine of inflamma-
respiratory infections was higher in intervention arm compared
tion and affects most cells and organs (51). In COPD IL-1 is
to placebo, 47% versus 35% and 40% versus 28% respectively.
increased in sputum, serum and BAL at stable state and exac-
Thus, the efficacy of CXCR2 inhibition was small and the poten-
erbation. IL-1 is primarily produced by macrophages, mono-
tial of increased adverse events has meant this target has not pro-
cytes and fibroblasts. IL-1A and IL-1B genes encode IL-1 α
gressed in COPD.
and IL-1 β, respectively (52). Both exert their effect by bind-
ing to IL-1 receptor 1 (IL-1R1), which is expressed in almost
Tumor necrosis factor –alpha (TNF-α) all cells. Unlike most other pro-inflammatory cytokines, IL-1
TNF-α is a potent pro-inflammatory mediator and plays a major exerts its effects at receptor and nucleus level. It stimulates local
role in driving inflammation in COPD exacerbations (43). The and systemic inflammation by facilitating the recruitment of
level of TNF-α in sputum rises significantly during an exacer- inflammatory cells. IL-1 antagonists have been used in treating
bation (42). It is mainly produced by monocytes and lympho- rheumatological conditions with mixed results and is associated
cytes. It is secreted in response to various stimuli. It exerts its with improved outcomes following myocardial infarction (52).
effect via TNF-α receptor 1 and TNF-α receptor 2 by activation Blocking the effects of IL-1 α and IL-1 β is thus a potential treat-
of nuclear factor-kappa B (NF-kB), which in turn leads to up ment strategy for COPD patients.
regulation of inflammatory genes. It stimulates leucocyte accu- Calverley and colleagues undertook a 1-year study of anti-
mulation and differentiation at the site of infection, as well as IL-1R1 in 324 subjects with COPD (53). The intervention arm
COPD: JOURNAL OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE 5

received a 600 mg loading of anti-IL-1 intravenously on day 1 difference was observed in health status. In a post hoc analysis
followed by 300 mg subcutaneous every 4 weeks for a total of those who had evidence of eosinophilic inflammation at screen-
14 doses. There was no reduction in exacerbation frequency ing, either a sputum eosinophil count >2% or a blood eosinophil
nor improvements in lung function and health status in those count >250 cells/μL, had a greater improvement in lung func-
receiving anti-IL-1R1 versus placebo. There were no differences tion, heath status and a numerical reduction in exacerbations.
in adverse events between the groups. In another study, 147 These findings perhaps suggesting greater efficacy in a more
patients with moderate to very severe COPD were randomized eosinophilic subgroup. There were no differences in adverse
to receive canakinumab, an IL1-1R1 antagonist or placebo for events between those receiving benralizumab versus placebo.
1 year. No statistical analysis was provided at the completion Dasgupta and colleagues undertook a small pilot study
of the study, but changes in lung function that are reported of mepolizumab in 18 COPD subjects (62). They found
were very similar between those receiving canalinumab versus that the sputum eosinophil count decreased in those receiv-
placebo (54). There were no deaths reported in the study and ing mepolizumab versus placebo, but there were no clinical
adverse events were similar between groups. No anti-IL-1 stud- improvements in terms of lung function or health status. Pavord
ies are currently ongoing. and colleagues undertook two recent phase III trials (METREX
Therefore, targeting neutrophilic inflammation via anti-IL-8 and METREO) in moderate to very severe COPD (46). Sub-
and anti-CXCR2 has led to small clinical benefit with evidence jects were randomized to receive 100 mg mepolizumab or
of reduced blood neutrophil counts and possibly increased risk placebo in 1:1 ratio (METREX, n = 836), 100 mg mepolizumab,
of infection. Targeting TNF-α and IL-1 has not been associ- 300 mg mepolizumab or placebo in 1:1:1 ratio (METREO, n
ated with efficacy and inhibiting TNF-α has led to substantial = 674) every 4 weeks for 52 weeks. Subject stratification into
and clinically important adverse events. These findings have eosinophilic or non-eosinophilic phenotypes were based on
stopped progress of biological therapy against pro-inflammatory blood eosinophil count of ࣙ150/µL on screening or ࣙ300/µL in
cytokines and neutrophilic inflammation in COPD. previous year and blood eosinophil count of <150/µL on screen-
ing or <300/µL in previous year, respectively. In METREX they
included eosinophilic and non-eosinophilic patients, whereas in
Anti- eosinophilic, T2 inflammation METREO they only included eosinophilic patients. In METREX
Biologics targeting T2-mediated inflammation has been suc- the mean annual rate of moderate or severe exacerbations in the
cessful in severe asthma and led to newly licensed therapies mepolizumab group was 1.40 per year compared to 1.71 per year
targeting IL-5 and its receptor (46,55–58). As described above in the placebo group (rate ratio, 0.82). In METREO, the mean
although eosinophilic inflammation is common in asthma it is annual rate of moderate or severe exacerbations was 1.19 per
less frequent in COPD and not associated with increased atopy. year in the 100-mg mepolizumab group and 1.27 per year in the
Thus, the mechanisms of the cause of eosinophilic inflamma- 300-mg mepolizumab group, compared to 1.49 per year in the
tion in COPD is poorly understood but does offer a potential placebo group (rate ratio [100 mg vs. placebo], 0.80; adjusted
therapeutic target. P = 0.07; rate ratio [300 mg vs. placebo], 0.86; adjusted P =
0.04). In a pre-planned post hoc analysis, those with eosinophilic
phenotype who were treated with 100 mg mepolizumab had an
Anti-IL- and anti-IL-R annual rate of moderate to severe exacerbation that was 18–20%
IL-5 is released by CD4+ T-helper cell type 2 (Th2) lym- lower than placebo group. Higher dose of mepolizumab did not
phocytes, innate lymphoid cells and eosinophils. Its receptor confer any additional benefit. The time to the first moderate or
Interleukin 5 receptor (IL-5R) is expressed by eosinophils and severe exacerbation was significantly longer with mepolizumab
basophil. IL-5 stimulates maturation and release of eosinophils than with placebo in the modified intention-to-treat population
from bone marrow and is an obligate survival factor for tis- with an eosinophilic phenotype (192 days versus 141 days; haz-
sue eosinophils (59). In asthma, biologics neutralizing IL- ard ratio, 0.75; adjusted P = 0.04). In keeping with the smaller
5 (mepolizumab and reslizumab) substantially reduce blood pilot study there were no improvements in lung function and
and sputum eosinophil counts and attenuate bronchial submu- health status in those receiving mepolizumab versus placebo and
cosal eosinophils by about 50% (60). The afucosylated mono- no differences in adverse events.
clonal antibody against the IL-5R, benralizumab, also induces Mepolizumab is the first biologic to demonstrate a reduction
antibody-mediated cell cytotoxicity and thus has a greater effect in COPD exacerbations. The findings from the mepolizumab
on reducing eosinophil number in the bronchial submucosa and benralizumab studies were consistent in showing that the
(61). magnitude of benefit was related to the intensity of eosinophilic
Brightling and colleagues undertook a study of anti-IL-5R inflammation (46). Importantly in both studies there was a
(benralizumab) in 101 patients with moderate to severe COPD poorer outcome in those with low blood eosinophil counts
(45). Subjects were randomized in a 1:1 ratio to either receive who received the biologic versus placebo. This is in contrast
benralizumab or placebo 4 weekly for the first 3 doses and to asthma where there is a failure of benefit in those with low
then 8 weekly for the remaining 5 doses. The primary outcome, eosinophil counts but no difference compared with placebo.
annual rate of acute exacerbations, was not met. The annual Whether anti-IL-5 or anti-IL-5R approaches increase risk of
rate of acute COPD exacerbations in placebo group was 0.92 exacerbations in COPD subjects with low eosinophil counts
and in intervention group was 0.95. The FEV1 was significantly needs to be further explored and the possible mechanisms
increased in those receiving benralizumab versus placebo but no elucidated.
6 A. YOUSUF AND C. E. BRIGHTLING

Anti-IL- been implicated in eosinophil recruitment to the airways and


IL-13 is an archetypal Th2 cytokine produced by T-cells, maturation in the bone marrow largely via its effects upon innate
mast cells and eosinophils and acts upon inflammatory lymphoid cells (76). IL-33 increased following experimental
cells and structural cells such as the bronchial epithe- cold in asthma and thus might play a role in the consequent
lium and airway smooth muscle (63). Anti-IL-13 signals inflammatory response and possible susceptibility to secondary
through interleukin 13 receptor type 1 (IL-13RI) and II bacterial infection in obstructive lung disease (77). The role of
with IL-13RI sharing the interleukin 4 (IL-4) receptor alpha the IL-33/ST2 axis in COPD is uncertain. A correlation between
chain subunit with the IL-4R interleukin 4 receptor (IL- blood eosinophil, smoking status and IL-33 in COPD has been
4R). Biologics have been tested in asthma that either neu- reported and IL-33 levels in peripheral blood and airways of
tralize IL-13 or inhibit IL-4Rα and thus inhibit both IL-4 COPD patients are increased (78). Both eosinophilic inflamma-
and −13. The former, lebrikizumab and tralokinumab, have tion and viral infection drive COPD exacerbations and therefore
consistently shown benefits compared to placebo with improved targeting the IL-33/ST2 axis might reduce COPD exacerbations.
lung function but effects upon exacerbations have been incon- Clinical trials of anti-IL33 or anti-ST2 biologics in COPD are
sistent and too small to warrant further development (64,65). In awaited.
contrast, anti-IL-4Rα has had more promising results in phase 3 Biologics might offer alternative strategies to target the expo-
in asthma (66). Of these three biologics, only lebrikizumab has some as well as the inflammatory response in the host. For exam-
been tested in COPD. The full results have not been reported ple, biologics can be developed to target bacteria; inhibit viru-
but the headline result in a press release reported that in COPD lence factors or block adhesion of viruses to cells and thus could
exacerbations were not reduced in those receiving lebrikizumab be adjuncts to antimicrobial strategies.
versus placebo.

Conclusion
Future biologics in COPD
The heterogeneity of COPD is beginning to be dissected and
In view of the poor response to biologics targeting neutrophilic our understanding of the interaction between the host, chronic
inflammation and pro-inflammatory cytokines versus the infection and other exposures is becoming clearer. This has
promising results in targeting eosinophilic inflammation atten- provided insights but also underscored the complexity of
tion in COPD is to initially explore other cytokines implicated these relationships overtime. Biological therapy in COPD has
in T2-mediated immunity and or eosinophilic inflammation. presented some surprises with respect to the lack of efficacy
Thymic stromal lymphopoietin (TSLP) is an IL-7 interleukin for treatments targeting neutrophilic inflammation and pro-
7 (IL-7) like cytokine, which signals via IL-7 receptor alpha (IL- inflammatory cytokines but has shown how blockade of these
7Rα) and thymic stromal lymphopoietin receptor (TSLPR) (67). cytokines can lead to profound increases in adverse events.
It stimulates thymocytes and B cell lymphopoiesis (68). TSLP This is contrasted with the promising responses to biologics
is produced by airway epithelial cells and stromal cells dur- targeting eosinophilic inflammation although responses are
ing inflammation (69). TSLP orchestrates response to epithe- more blunted than observed in asthma. These differences
lial injury by acting on CD4, CD8 T cells, B cells, mast cells, are possibly due to different underlying mechanisms driving
basophils, eosinophils and innate lymphoid cells (70). There the eosinophilic inflammation between asthma and COPD but
has been a great deal of interest in role of TSLP in pathogen- are pointing towards the possibility of biological therapy for
esis of asthma and COPD due to its ability to activate pro- a subgroup with persistent eosinophilic inflammation. This
inflammatory cells. Studies have shown increased expression of will perhaps present other opportunities for biologics targeting
TSLP in airways disease (71,72). Tezepelumab a monoclonal more upstream molecules such as TSLP and IL-33. Biologics
antibody that binds to TSLP and prevents it from attaching to might also offer alternative strategies to targeting acute and
TSLP receptor reduced asthma exacerbations, improved lung chronic infection, which is problem in many subjects with
function and asthma control (73). It attenuated the peripheral COPD. Biologics have not yet entered the clinic for COPD but
blood eosinophil count although response was also observed the likelihood is that the wait will be not much longer and then
in those without an elevated blood eosinophil count. Whether we shall need to better understand which biological therapy has
targeting TSLP has benefit in an unselected COPD population value in which COPD patient to improve what outcome.
or in an eosinophilic subgroup is unknown and warrants fur-
ther investigation. This reduction in exacerbation frequency was
irrespective of eosinophil count. There has not been a similar Declaration of interest
study on the effect of anti-TSLP in COPD. None.
There is also increasing interest in IL-33 as a target particu-
larly in asthma. IL-33 is an alarmin released from the epithe-
References
lium following damage (74). IL-33 is an IL-1 family alarmin
cytokine constitutively expressed at epithelial barrier surfaces 1. Global Strategy for the Diagnosis, Management and Preven-
where it is rapidly released from cells during tissue injury. IL- tion of COPD, Global Initiative for Chronic Obstructive Lung
33 signals through a receptor complex of IL-1 receptor-like 1 Disease (GOLD) 2017 [cited 2017 02/10/2017]. Available from:
http://goldcopd.org.
(IL1RL1) (known as ST2) and IL-1 receptor accessory protein 2. Lelieveld J, Evans JS, Fnais M, Giannadaki D, Pozzer A. The contribu-
(IL1RAcP) to initiate Myeloid differentiation primary response tion of outdoor air pollution sources to premature mortality on a global
88 (MyD88)-dependent inflammatory pathways (75). IL-33 has scale. Nature. 2015;525(7569):367–71. doi:10.1038/nature15371.
COPD: JOURNAL OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE 7

3. Goren A, Gupta S, Dong P, Feng Y, Chen C, Liu D. Burden biopsies in asthma and chronic bronchitis. Am J Respir Crit Care Med.
of smoking among adults with COPD, chronic bronchitis, and 1997;156(2 Pt 1):591–9. doi:10.1164/ajrccm.156.2.9609066.
emphysema in urban China. Int J Clin Pract. 2015;69(9):1015–28. 23. Polosa R, Prosperini G, Leir SH, Holgate ST, Lackie PM, Davies
doi:10.1111/ijcp.12680. DE. Expression of c-erbB receptors and ligands in human bronchial
4. The global burden of disease [Report]. [cited 2017 12/12/2017]. Avail- mucosa. Am J Respir Cell Mol Biol. 1999;20(5):914–23.
able from: http://www.who.int/respiratory/copd/en/. 24. O’Donnell RA, Richter A, Ward J, Angco G, Mehta A, Rousseau
5. Adeloye D, Chua S, Lee C, Basquill C, Papana A, Theodoratou K, et al. Expression of ErbB receptors and mucins in the air-
E, et al. Global and regional estimates of COPD prevalence: Sys- ways of long term current smokers. Thorax. 2004;59(12):1032–40.
tematic review and meta-analysis. J Glob Health. 2015;5(2):020415. doi:10.1136/thx.2004.028043.
doi:10.7189/jogh.05-020415. 25. de Boer WI, Hau CM, van Schadewijk A, Stolk J, van Krieken JH,
6. The Burden of Lung Disease: a statistics report from the British Hiemstra PS. Expression of epidermal growth factors and their recep-
Thoracic Society 2006 [updated 2006]. Available from: https://www. tors in the bronchial epithelium of subjects with chronic obstruc-
brit-thoracic.org.uk/document-library/delivery-of-respiratory- tive pulmonary disease. Am J Clin Pathol. 2006;125(2):184–92.
care/burden-of-lung-disease/burden-of-lung-disease-2006/. doi:10.1309/W1AX-KGT7-UA37-X257.
7. Castaldi PJ, Benet M, Petersen H, Rafaels N, Finigan J, Paoletti M, 26. Kim S, Lewis C, Nadel JA. CCL20/CCR6 feedback exaggerates epi-
et al. Do COPD subtypes really exist? COPD heterogeneity and clus- dermal growth factor receptor-dependent MUC5AC mucin pro-
tering in 10 independent cohorts. Thorax. 2017;72(11):998–1006. duction in human airway epithelial (NCI-H292) cells. J Immunol.
doi:10.1136/thoraxjnl-2016-209846. 2011;186(6):3392–400. doi:10.4049/jimmunol.1003377.
8. Di Stefano A, Capelli A, Lusuardi M, Balbo P, Vecchio C, Maestrelli 27. Bosken CH, Wiggs BR, Pare PD, Hogg JC. Small airway dimen-
P, et al. Severity of airflow limitation is associated with severity sions in smokers with obstruction to airflow. Am Rev Respir Dis.
of airway inflammation in smokers. Am J Respir Crit Care Med. 1990;142(3):563–70. doi:10.1164/ajrccm/142.3.563.
1998;158(4):1277–85. doi:10.1164/ajrccm.158.4.9802078. 28. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L, Mapp
9. Barnes PJ. Cellular and molecular mechanisms of asthma and COPD. CE, et al. CD8+ T-lymphocytes in peripheral airways of smokers with
Clin Sci (Lond). 2017;131(13):1541–58. doi:10.1042/CS20160487. chronic obstructive pulmonary disease. Am J Respir Crit Care Med.
10. Eapen MS, Myers S, Walters EH, Sohal SS. Airway inflam- 1998;157(3 Pt 1):822–6. doi:10.1164/ajrccm.157.3.9709027.
mation in chronic obstructive pulmonary disease (COPD): 29. Araya J, Cambier S, Markovics JA, Wolters P, Jablons D, Hill A, et al.
a true paradox. Expert Rev Respir Med. 2017;11(10):827–39. Squamous metaplasia amplifies pathologic epithelial-mesenchymal
doi:10.1080/17476348.2017.1360769. interactions in COPD patients. J Clin Invest. 2007;117(11):3551–62.
11. Finney LJ, Ritchie A, Pollard E, Johnston SL, Mallia P. Lower air- doi:10.1172/JCI32526.
way colonization and inflammatory response in COPD: a focus 30. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk J, Hiem-
on Haemophilus influenzae. Int J Chron Obstruct Pulmon Dis. stra PS, et al. Transforming growth factor beta1 and recruitment of
2014;9:1119–32. doi:10.2147/COPD.S54477. macrophages and mast cells in airways in chronic obstructive pul-
12. George L, Brightling CE. Eosinophilic airway inflammation: role in monary disease. Am J Respir Crit Care Med. 1998;158(6):1951–7.
asthma and chronic obstructive pulmonary disease. Ther Adv Chronic doi:10.1164/ajrccm.158.6.9803053.
Dis. 2016;7(1):34–51. doi:10.1177/2040622315609251. 31. Takizawa H, Tanaka M, Takami K, Ohtoshi T, Ito K, Satoh M, et al.
13. Saetta M, Di Stefano A, Maestrelli P, Turato G, Ruggieri MP, Rog- Increased expression of transforming growth factor-beta1 in small
geri A, et al. Airway eosinophilia in chronic bronchitis during exac- airway epithelium from tobacco smokers and patients with chronic
erbations. Am J Respir Crit Care Med. 1994;150(6 Pt 1):1646–52. obstructive pulmonary disease (COPD). Am J Respir Crit Care Med.
doi:10.1164/ajrccm.150.6.7952628. 2001;163(6):1476–83. doi:10.1164/ajrccm.163.6.9908135.
14. Papi ABC, Pedersen S, Reddel HK. Asthma. The Lancet. 2017. Epub 32. Kranenburg AR, Willems-Widyastuti A, Moori WJ, Sterk PJ, Alagap-
19/12/2017. pan VK, de Boer WI, et al. Enhanced bronchial expression of extracel-
15. Saetta M, Turato G, Maestrelli P, Mapp CE, Fabbri LM. Cel- lular matrix proteins in chronic obstructive pulmonary disease. Am J
lular and structural bases of chronic obstructive pulmonary Clin Pathol. 2006;126(5):725–35.
disease. Am J Respir Crit Care Med. 2001;163(6):1304–9. 33. Stockley RA. Neutrophils and the pathogenesis of COPD. Chest.
doi:10.1164/ajrccm.163.6.2009116. 2002;121(5 Suppl):151S–5S.
16. Maestrelli P, Saetta M, Mapp CE, Fabbri LM. Remodeling in response 34. Turino GM, Seniorrm, Garg BD, Keller S, Levi MM, Mandl I.
to infection and injury. Airway inflammation and hypersecretion Serum elastase inhibitor deficiency and alpha 1-antitrypsin deficiency
of mucus in smoking subjects with chronic obstructive pulmonary in patients with obstructive emphysema. Science. 1969;165(3894):
disease. Am J Respir Crit Care Med. 2001;164(10 Pt 2):S76–80. 709–11.
doi:10.1164/ajrccm.164.supplement_2.2106067. 35. Lacoste JY, Bousquet J, Chanez P, Van Vyve T, Simony-Lafontaine
17. Hogg JC. Pathophysiology of airflow limitation in chronic J, Lequeu N, et al. Eosinophilic and neutrophilic inflammation in
obstructive pulmonary disease. Lancet. 2004;364(9435):709–21. asthma, chronic bronchitis, and chronic obstructive pulmonary dis-
doi:10.1016/S0140-6736(04)16900-6. ease. J Allergy Clin Immunol. 1993;92(4):537–48.
18. Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, 36. Brightling CE, Monteiro W, Ward R, Parker D, Morgan MD, Ward-
et al. The nature of small-airway obstruction in chronic obstruc- law AJ, et al. Sputum eosinophilia and short-term response to pred-
tive pulmonary disease. N Engl J Med. 2004;350(26):2645–53. nisolone in chronic obstructive pulmonary disease: a randomised
doi:10.1056/NEJMoa032158. controlled trial. Lancet. 2000;356(9240):1480–5. doi:10.1016/S0140-
19. McDonough JE, Yuan R, Suzuki M, Seyednejad N, Elliott 6736(00)02872-5.
WM, Sanchez PG, et al. Small-airway obstruction and emphy- 37. Brightling CE, McKenna S, Hargadon B, Birring S, Green R, Siva R,
sema in chronic obstructive pulmonary disease. N Engl J Med. et al. Sputum eosinophilia and the short term response to inhaled
2011;365(17):1567–75. doi:10.1056/NEJMoa1106955. mometasone in chronic obstructive pulmonary disease. Thorax.
20. Rennard SI, Togo S, Holz O. Cigarette smoke inhibits alveolar repair: a 2005;60(3):193–8. doi:10.1136/thx.2004.032516.
mechanism for the development of emphysema. Proc Am Thorac Soc. 38. Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K, Gleich GJ,
2006;3(8):703–8. doi:10.1513/pats.200605-121SF. Berman L, et al. Sputum eosinophilia predicts benefit from prednisone
21. Wang H, Liu X, Umino T, Skold CM, Zhu Y, Kohyama T, in smokers with chronic obstructive bronchitis. Am J Respir Crit Care
et al. Cigarette smoke inhibits human bronchial epithelial cell Med. 1998;158(5 Pt 1):1511–7. doi:10.1164/ajrccm.158.5.9804028.
repair processes. Am J Respir Cell Mol Biol. 2001;25(6):772–9. 39. Green RH, Brightling CE, McKenna S, Hargadon B, Parker D,
doi:10.1165/ajrcmb.25.6.4458. Bradding P, et al. Asthma exacerbations and sputum eosinophil
22. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace E, Rizzo counts: a randomised controlled trial. Lancet. 2002;360(9347):
A, et al. Transforming growth factor-beta expression in mucosal 1715–21. doi:10.1016/S0140-6736(02)11679-5.
8 A. YOUSUF AND C. E. BRIGHTLING

40. Bafadhel M, McKenna S, Terry S, Mistry V, Pancholi M, Venge P, with elevated blood eosinophil counts: results from two multicen-
et al. Blood eosinophils to direct corticosteroid treatment of exac- tre, parallel, double-blind, randomised, placebo-controlled, phase
erbations of chronic obstructive pulmonary disease: a randomized 3 trials. Lancet Respir Med. 2015;3(5):355–66. doi:10.1016/S2213-
placebo-controlled trial. Am J Respir Crit Care Med. 2012;186(1): 2600(15)00042-9.
48–55. doi:10.1164/rccm.201108-1553OC. 58. Yancey SW, Ortega HG, Keene ON, Mayer B, Gunsoy NB, Brightling
41. Bafadhel M, Davies L, Calverley PM, Aaron SD, Brightling CE, CE, et al. Meta-analysis of asthma-related hospitalization in
Pavord ID. Blood eosinophil guided prednisolone therapy for exacer- mepolizumab studies of severe eosinophilic asthma. J Allergy Clin
bations of COPD: a further analysis. Eur Respir J. 2014;44(3):789–91. Immunol. 2017;139(4):1167–75 e2. doi:10.1016/j.jaci.2016.08.008.
doi:10.1183/09031936.00062614. 59. Bagnasco D, Ferrando M, Varricchi G, Puggioni F, Passalacqua G,
42. Aaron SD, Vandemheen KL, Maltais F, Field SK, Sin DD, Bourbeau Canonica GW. Anti-Interleukin 5 (IL-5) and IL-5Ra biological drugs:
J, et al. TNFalpha antagonists for acute exacerbations of COPD: a efficacy, safety, and future perspectives in severe eosinophilic asthma.
randomised double-blind controlled trial. Thorax. 2013;68(2):142–8. Front Med (Lausanne). 2017;4:135. doi:10.3389/fmed.2017.00135.
doi:10.1136/thoraxjnl-2012-202432. 60. Cabon Y, Molinari N, Marin G, Vachier I, Gamez AS, Chanez
43. Rennard SI, Fogarty C, Kelsen S, Long W, Ramsdell J, Allison J, P, et al. Comparison of anti-interleukin-5 therapies in patients
et al. The safety and efficacy of infliximab in moderate to severe with severe asthma: global and indirect meta-analyses of random-
chronic obstructive pulmonary disease. Am J Respir Crit Care Med. ized placebo-controlled trials. Clin Exp Allergy. 2017;47(1):129–38.
2007;175(9):926–34. doi:10.1164/rccm.200607-995OC. doi:10.1111/cea.12853.
44. Kirsten AM, Forster K, Radeczky E, Linnhoff A, Balint B, Watz H, et al. 61. Saco TV, Pepper AN, Lockey RF. Benralizumab for the treat-
The safety and tolerability of oral AZD5069, a selective CXCR2 antag- ment of asthma. Expert Rev Clin Immunol. 2017;13(5):405–13.
onist, in patients with moderate-to-severe COPD. Pulm Pharmacol doi:10.1080/1744666X.2017.1316194.
Ther. 2015;31:36–41. doi:10.1016/j.pupt.2015.02.001. 62. Dasgupta A, Kjarsgaard M, Capaldi D, Radford K, Aleman F, Boy-
45. Brightling CE, Bleecker ER, Panettieri RA, Jr., Bafadhel M, She D, lan C, et al. A pilot randomised clinical trial of mepolizumab
Ward CK, et al. Benralizumab for chronic obstructive pulmonary dis- in COPD with eosinophilic bronchitis. Eur Respir J. 2017;49(3).
ease and sputum eosinophilia: a randomised, double-blind, placebo- doi:10.1183/13993003.02486-2016.
controlled, phase 2a study. Lancet Respir Med. 2014;2(11):891–901. 63. Brightling CE, Saha S, Hollins F. Interleukin-13: prospects for new
doi:10.1016/S2213-2600(14)70187-0. treatments. Clin Exp Allergy. 2010;40(1):42–9. doi:10.1111/j.1365-
46. Pavord ID, Chanez P, Criner GJ, Kerstjens HAM, Korn S, 2222.2009.03383.x.
Lugogo N, et al. Mepolizumab for Eosinophilic Chronic Obstruc- 64. Hanania NAKP, Chapman KR, Bateman ED, Kopecky P, Paggiaro
tive Pulmonary Disease. N Engl J Med. 2017;377(17):1613–29. P, et al. Efficacy and safety of lebrikizumab in patients with uncon-
doi:10.1056/NEJMoa1708208. trolled asthma (LAVOLTA I and LAVOLTA II): replicate, phase 3, ran-
47. Mahler DA, Huang S, Tabrizi M, Bell GM. Efficacy and safety of a mon- domised, double-blind, placebo-controlled trials. Lancet Respir Med.
oclonal antibody recognizing interleukin-8 in COPD: a pilot study. 2016;4:781–96.
Chest. 2004;126(3):926–34. doi:10.1378/chest.126.3.926. 65. Brightling CECP, Leight R, et al. Efficacy and safety of tralokinumab in
48. Brightling C, Berry M, Amrani Y. Targeting TNF-alpha: a novel ther- patients with severe uncontrolled asthma: a randomised, double-blind,
apeutic approach for asthma. J Allergy Clin Immunol. 2008;121(1):5– placebo-controlled, phase 2b trial. The Lancet. 2015;3(9):629–701.
10; quiz 1–2. doi:10.1016/j.jaci.2007.10.028. 66. Wenzel S, Castro M, Corren J, Maspero J, Wang L, Zhang B, et al.
49. Suissa S, Ernst P, Hudson M. TNF-alpha antagonists and Dupilumab efficacy and safety in adults with uncontrolled per-
the prevention of hospitalisation for chronic obstructive pul- sistent asthma despite use of medium-to-high-dose inhaled corti-
monary disease. Pulm Pharmacol Ther. 2008;21(1):234–8. costeroids plus a long-acting beta2 agonist: a randomised double-
doi:10.1016/j.pupt.2007.03.003. blind placebo-controlled pivotal phase 2b dose-ranging trial. Lancet.
50. van der Vaart H, Koeter GH, Postma DS, Kauffman HF, ten 2016;388(10039):31–44. doi:10.1016/S0140-6736(16)30307-5.
Hacken NH. First study of infliximab treatment in patients with 67. Yadava K, Massacand J, Mosconi I, Nicod LP, Harris NL, Mars-
chronic obstructive pulmonary disease. Am J Respir Crit Care Med. land BJ. Thymic stromal lymphopoietin plays divergent roles in
2005;172(4):465–9. doi:10.1164/rccm.200501-147OC. murine models of atopic and nonatopic airway inflammation. Allergy.
51. Dinarello CA, Simon A, van der Meer JW. Treating inflammation by 2014;69(10):1333–42. doi:10.1111/all.12469.
blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug 68. Ziegler SF, Roan F, Bell BD, Stoklasek TA, Kitajima M, Han H. The
Discov. 2012;11(8):633–52. doi:10.1038/nrd3800. biology of thymic stromal lymphopoietin (TSLP). Adv Pharmacol.
52. Gabay C, Lamacchia C, Palmer G. IL-1 pathways in inflamma- 2013;66:129–55. doi:10.1016/B978-0-12-404717-4.00004-4.
tion and human diseases. Nat Rev Rheumatol. 2010;6(4):232–41. 69. Lloyd CM, Saglani S. Epithelial cytokines and pulmonary
doi:10.1038/nrrheum.2010.4. allergic inflammation. Curr Opin Immunol. 2015;34:52–8.
53. Calverley PMA, Sethi S, Dawson M, Ward CK, Finch DK, Penney doi:10.1016/j.coi.2015.02.001.
M, et al. A randomised, placebo-controlled trial of anti-interleukin- 70. Redhu NS, Gounni AS. Function and mechanisms of TSLP/TSLPR
1 receptor 1 monoclonal antibody MEDI8968 in chronic obstructive complex in asthma and COPD. Clin Exp Allergy. 2012;42(7):994–
pulmonary disease. Respir Res. 2017;18(1):153. doi:10.1186/s12931- 1005. doi:10.1111/j.1365-2222.2011.03919.x.
017-0633-7. 71. Ying S, O’Connor B, Ratoff J, Meng Q, Fang C, Cousins D, et al. Expres-
54. Safety and Efficacy of Multiple Doses of Canakinumab (ACZ885) sion and cellular provenance of thymic stromal lymphopoietin and
in Chronic Obstructive Pulmonary Disease (COPD) Patients [Inter- chemokines in patients with severe asthma and chronic obstructive
net]. 2017 [cited 12/11/2017]. Available from: https://clinicaltrials.gov/ pulmonary disease. J Immunol. 2008;181(4):2790–8.
ct2/show/NCT00581945?term=NCT00581945&rank=1. 72. Shikotra A, Choy DF, Ohri CM, Doran E, Butler C, Hargadon B,
55. Ortega HG, Liu MC, Pavord ID, Brusselle GG, FitzGerald JM, et al. Increased expression of immunoreactive thymic stromal lym-
Chetta A, et al. Mepolizumab treatment in patients with severe phopoietin in patients with severe asthma. J Allergy Clin Immunol.
eosinophilic asthma. N Engl J Med. 2014;371(13):1198–207. 2012;129(1):104–11 e1–9. doi:10.1016/j.jaci.2011.08.031.
doi:10.1056/NEJMoa1403290. 73. Corren J, Parnes JR, Wang L, Mo M, Roseti SL, Griffiths JM, et al.
56. Bel EH, Wenzel SE, Thompson PJ, Prazma CM, Keene ON, Tezepelumab in Adults with Uncontrolled Asthma. N Engl J Med.
Yancey SW, et al. Oral glucocorticoid-sparing effect of mepolizumab 2017;377(10):936–46. doi:10.1056/NEJMoa1704064.
in eosinophilic asthma. N Engl J Med. 2014;371(13):1189–97. 74. Kaur D, Gomez E, Doe C, Berair R, Woodman L, Saunders R, et al.
doi:10.1056/NEJMoa1403291. IL-33 drives airway hyper-responsiveness through IL-13-mediated
57. Castro M, Zangrilli J, Wechsler ME, Bateman ED, Brusselle GG, mast cell: airway smooth muscle crosstalk. Allergy. 2015;70(5):556–
Bardin P, et al. Reslizumab for inadequately controlled asthma 67. doi:10.1111/all.12593.
COPD: JOURNAL OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE 9

75. Cayrol C, Girard JP. Interleukin-33 (IL-33): a nuclear cytokine ing rhinovirus-induced asthma exacerbations in vivo. Am J Respir
from the IL-1 family. Immunol Rev. 2018;281(1):154–68. Crit Care Med. 2014;190(12):1373–82. doi:10.1164/rccm.201406-
doi:10.1111/imr.12619. 1039OC.
76. Cayrol C, Girard JP. IL-33: an alarmin cytokine with crucial roles 78. Xia J, Zhao J, Shang J, Li M, Zeng Z, Zhao J, et al. Increased
in innate immunity, inflammation and allergy. Curr Opin Immunol. IL-33 expression in chronic obstructive pulmonary disease.
2014;31:31–7. doi:10.1016/j.coi.2014.09.004. Am J Physiol Lung Cell Mol Physiol. 2015;308(7):L619–27.
77. Jackson DJ, Makrinioti H, Rana BM, Shamji BW, Trujillo-Torralbo doi:10.1152/ajplung.00305.2014.
MB, Footitt J, et al. IL-33-dependent type 2 inflammation dur-

You might also like