+ All Categories
Home > Documents > Targeting Immune Pathways for Therapy in Asthma and Chronic Obstructive Pulmonary Disease

Targeting Immune Pathways for Therapy in Asthma and Chronic Obstructive Pulmonary Disease

Date post: 25-Nov-2023
Category:
Upload: independent
View: 0 times
Download: 0 times
Share this document with a friend
7
TRANSATLANTIC AIRWAY CONFERENCE Targeting Immune Pathways for Therapy in Asthma and Chronic Obstructive Pulmonary Disease Guy Brusselle 1,2 and Ken Bracke 1 1 Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium; and 2 Departments of Epidemiology and Respiratory Medicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands Abstract Asthma and chronic obstructive pulmonary disease (COPD) are highly prevalent chronic inammatory diseases of the airways, with differences in etiology, pathogenesis, immunologic mechanisms, clinical presentation, comorbidities, prognosis, and response to treatment. In mild to moderate early-onset allergic asthma, the Th2-driven eosinophilic airway inammation and the ensuing disease can be well controlled with maintenance treatment with inhaled corticosteroids (ICS). In real-life settings, asthma control can be improved by facilitating adherence to ICS treatment and by optimizing inhaler technique. In patients with uncontrolled severe asthma, old and novel therapies targeting specic immunologic pathways should be added according to the underlying endotype/ phenotype. In COPD, there is a high unmet need for safe and effective antiinammatory treatments that not only prevent exacerbations but also have a benecial impact on the course of the disease and improve survival. Although several new approaches aim to target the chronic neutrophilic pulmonary inammation per se in patients with COPD, strategies that target the underlying causes of the pulmonary neutrophilia (e.g., smoking, chronic infection, and oxidative stress) might be more successful. In both chronic airway diseases (especially in more difcult, complex cases), the choice of the optimal treatment should be based not only on arbitrary clinical labels but also on the underlying immunopathology. Keywords: asthma; chronic obstructive pulmonary disease; phenotypes; treatment; monoclonal antibodies (Received in original form March 18, 2014; accepted in final form June 17, 2014 ) Although G.B. is currently guideline director of the European Respiratory Society, this manuscript only reflects his personal opinions and ideas. Correspondence and requests for reprints should be addressed to Guy Brusselle, M.D., Ph.D., Ghent University Hospital, Respiratory Medicine, De Pintelaan 185, B-9000 Ghent, Belgium. E-mail: [email protected] Ann Am Thorac Soc Vol 11, Supplement 5, pp S322–S328, Dec 2014 Copyright © 2014 by the American Thoracic Society DOI: 10.1513/AnnalsATS.201403-118AW Internet address: www.atsjournals.org Asthma and chronic obstructive pulmonary disease (COPD) are chronic inammatory diseases of the airways, which share clinical symptoms (shortness of breath, wheezing, coughing, and sputum production), pathophysiological mechanisms (airow limitation and bronchial hyperresponsiveness), and pathogenetic mechanisms (i.e., interactions between environmental exposures and genetic susceptibility) (1). Although asthma has been classically considered as caused by an allergic eosinophilic airway inammation, most frequently starting in early life (childhood and adolescence), it has become evident that asthma is a very heterogeneous syndrome, encompassing other phenotypes of asthma, such as nonallergic asthma, noneosinophilic asthma (characterized by neutrophilic or paucigranulocytic airway inammation), and asthma that starts only in adulthood (adult-onset/late-onset asthma) (24). Unfortunately, some patients with asthma do smoke and thereby respond less well to maintenance treatment with inhaled corticosteroids (ICS). At the other end of the spectrum of chronic airway disease, COPD has been classically dened as an irreversible airow limitation in smokers or ex-smokers, which is associated with chronic neutrophilic inammation of the small airways (bronchiolitis) and progressive destruction of the lung parenchyma (emphysema) (5). However, COPD is also a heterogeneous disease that encompasses multiple phenotypes (e.g., airway-predominant vs. emphysema-predominant phenotypes) and can occur in never-smokers (caused by exposure to biomass fuel smoke, outdoor air pollution, and/or occupational exposures) (6, 7). Moreover, the airow limitation is partially reversible in approximately one-half of the patients with COPD (8). In this opinion paper, we focus on asthma in nonsmoking adults and on COPD in (ex)smokers, because most randomized clinical trials (RCTs) systematically exclude current smokers (or ex-smokers with a smoking history of .10 pack-years) in trials of asthma, and never-smokers (or smokers with a smoking history of ,10 pack-years) in trials of COPD. Apparently, for pharmaceutical companies and regulators, the magic number of 10 (pack-years of smoking) is the critical cut-off point for S322 AnnalsATS Volume 11 Supplement 5 | December 2014
Transcript

TRANSATLANTIC AIRWAYCONFERENCE

Targeting Immune Pathways for Therapy in Asthma and ChronicObstructive Pulmonary DiseaseGuy Brusselle1,2 and Ken Bracke1

1Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium; and 2Departments of Epidemiology and RespiratoryMedicine, Erasmus Medical Center Rotterdam, Rotterdam, The Netherlands

Abstract

Asthma and chronic obstructive pulmonary disease (COPD) arehighly prevalent chronic inflammatory diseases of the airways, withdifferences in etiology, pathogenesis, immunologic mechanisms,clinical presentation, comorbidities, prognosis, and response totreatment. In mild to moderate early-onset allergic asthma, theTh2-driven eosinophilic airway inflammation and the ensuingdisease can be well controlled with maintenance treatment withinhaled corticosteroids (ICS). In real-life settings, asthma controlcan be improved by facilitating adherence to ICS treatment andby optimizing inhaler technique. In patients with uncontrolled severeasthma, old and novel therapies targeting specific immunologicpathways should be added according to the underlying endotype/

phenotype. InCOPD, there is a high unmet need for safe and effectiveantiinflammatory treatments that not only prevent exacerbationsbut also have a beneficial impact on the course of the disease andimprove survival. Although several new approaches aim to target thechronic neutrophilic pulmonary inflammation per se in patientswith COPD, strategies that target the underlying causes of thepulmonary neutrophilia (e.g., smoking, chronic infection, andoxidative stress) might be more successful. In both chronic airwaydiseases (especially in more difficult, complex cases), the choiceof the optimal treatment should be based not only on arbitraryclinical labels but also on the underlying immunopathology.

Keywords: asthma; chronic obstructive pulmonary disease;phenotypes; treatment; monoclonal antibodies

(Received in original form March 18, 2014; accepted in final form June 17, 2014 )

Although G.B. is currently guideline director of the European Respiratory Society, this manuscript only reflects his personal opinions and ideas.

Correspondence and requests for reprints should be addressed to Guy Brusselle, M.D., Ph.D., Ghent University Hospital, Respiratory Medicine, De Pintelaan185, B-9000 Ghent, Belgium. E-mail: [email protected]

Ann Am Thorac Soc Vol 11, Supplement 5, pp S322–S328, Dec 2014Copyright © 2014 by the American Thoracic SocietyDOI: 10.1513/AnnalsATS.201403-118AWInternet address: www.atsjournals.org

Asthma and chronic obstructive pulmonarydisease (COPD) are chronic inflammatorydiseases of the airways, which shareclinical symptoms (shortness of breath,wheezing, coughing, and sputum production),pathophysiological mechanisms (airflowlimitation and bronchial hyperresponsiveness),and pathogenetic mechanisms (i.e.,interactions between environmentalexposures and genetic susceptibility) (1).Although asthma has been classicallyconsidered as caused by an allergiceosinophilic airway inflammation, mostfrequently starting in early life (childhoodand adolescence), it has become evidentthat asthma is a very heterogeneoussyndrome, encompassing other phenotypesof asthma, such as nonallergic asthma,noneosinophilic asthma (characterized by

neutrophilic or paucigranulocytic airwayinflammation), and asthma that startsonly in adulthood (adult-onset/late-onsetasthma) (2–4). Unfortunately, somepatients with asthma do smoke and therebyrespond less well to maintenance treatmentwith inhaled corticosteroids (ICS).

At the other end of the spectrum ofchronic airway disease, COPD has beenclassically defined as an irreversible airflowlimitation in smokers or ex-smokers, whichis associated with chronic neutrophilicinflammation of the small airways(bronchiolitis) and progressive destructionof the lung parenchyma (emphysema) (5).However, COPD is also a heterogeneousdisease that encompasses multiplephenotypes (e.g., airway-predominant vs.emphysema-predominant phenotypes)

and can occur in never-smokers (causedby exposure to biomass fuel smoke,outdoor air pollution, and/or occupationalexposures) (6, 7). Moreover, the airflowlimitation is partially reversible inapproximately one-half of the patientswith COPD (8). In this opinion paper,we focus on asthma in nonsmokingadults and on COPD in (ex)smokers,because most randomized clinical trials(RCTs) systematically exclude currentsmokers (or ex-smokers with a smokinghistory of .10 pack-years) in trials ofasthma, and never-smokers (or smokerswith a smoking history of ,10 pack-years)in trials of COPD. Apparently, forpharmaceutical companies and regulators,the magic number of 10 (pack-years ofsmoking) is the critical cut-off point for

S322 AnnalsATS Volume 11 Supplement 5| December 2014

the differential diagnosis between asthmaand COPD.

Asthma

Asthma is characterized by chronic airwayinflammation, which is associated withbronchial hyperresponsiveness, leading tovariable airflow limitation and respiratorysymptoms (cough, wheezing, chesttightness, and/or shortness of breath) onexposure to specific allergens or nonspecifictriggers. In the past decades, asthma hasbeen considered as a homogeneous diseasecaused by an allergen-induced, Th2-driveneosinophilic airway inflammation, whichhas been modeled extensively in mice usingovalbumin or house dust mite as allergens(9, 10). However, clinical and translationalresearch in recent years has demonstratedthat asthma is a heterogeneous diseaseencompassing many different endotypesand phenotypes (Figure 1) (2, 3). Althoughseveral classification schemes of asthmahave been proposed, for the purpose ofthis paper we will focus on the differentinflammatory phenotypes of asthma,encompassing eosinophilic asthma,neutrophilic asthma, mixed granulocyticasthma, and paucigranulocytic asthma.Importantly, whereas allergy has beenregarded as the main driver in asthmapathogenesis, it has become clear thata large proportion of adult patients,even those with eosinophilic asthma,are nonallergic (defined by negative skinprick tests and serum allergen-specificradioallergosorbent tests) (11, 12).

The current mainstay treatment ofpersistent asthma is inhaled corticosteroids(ICS), with or without long-acting b2-agonists (LABA). Treatment with ICS for1 month or more significantly reduces thepathological signs of airway inflammationin asthma, decreasing the number ofTh2 lymphocytes and eosinophils andimproving progressively the bronchialhyperresponsiveness with prolongedtreatment (13, 14). Classical RCTs haveprovided overwhelming evidence of theefficacy and safety of ICS in children andadults with persistent asthma, leading towell-controlled disease in the majorityof patients with mild to moderate asthma.However, observational real-life studiesthroughout the world have shownthat many patients with asthma haveuncontrolled disease due to a combination

of factors: mainly noncompliance withmaintenance treatment (ICS), incorrectinhalation technique, the presence ofcomorbidities (e.g., chronic rhinosinusitis,gastroesophageal reflux, obesity, obstructivesleep apnea syndrome, and depression),and persistent exposure to allergens and/orirritants (e.g., cigarette smoke) (15, 16).The implication of this discrepancy isthat—at the population level—major gainsin asthma control could be obtained byfacilitating adherence to chronic ICStreatment and by optimizing inhalationdevices and techniques (17). However,a subgroup of patients with refractorysevere asthma remain uncontrolled despiteoptimal adherence to treatment with high-dose ICS and even oral corticosteroids,implicating a high unmet medical need.

Although ICS are an efficaciousantiinflammatory treatment in patientswith mild to moderate asthma, they do notmodify the course of the disease and thusdo not cure asthma. In the past decade,several large RCTs and Cochrane reviewshave proven that (subcutaneous) specificimmunotherapy with the appropriateallergen is efficacious and safe in well-selected patients with allergic rhinitis.Due to the increased risk of severeallergic reactions, including anaphylaxisand severe asthma attacks, specific allergenimmunotherapy has been contraindicatedin patients with uncontrolled asthma.

Targeting specific immune pathways isbecoming an attractive approach in patientswith uncontrolled severe asthma (9).Because severe asthma is a heterogeneoussyndrome, the specific immunologicadd-on therapies (on top of high-doseICS1LABA) should be targeted to theappropriate severe asthma phenotype(18). The classic example is the anti-IgEmonoclonal antibody omalizumab, whichis indicated in patients with severe allergicasthma with frequent exacerbations andpersistent airflow limitation. Omalizumabhas been shown to attenuate eosinophilicairway inflammation in subjects withallergic asthma and to reduce the frequencyof exacerbations in severe allergic asthma(19). Moreover, an increased fractionalexcretion of nitric oxide in the exhaledair, an increased blood eosinophilia,and increased serum levels of periostin,a protein produced by airway epithelial cellsunder the influence of the Th2-cytokineIL-13, are predictors of response totreatment with omalizumab (20). In

patients with refractory eosinophilicasthma, add-on treatment with the anti–IL-5 monoclonal antibody mepolizumabhas been shown to significantly reduce therate of severe asthma exacerbations (11,21). Importantly, approximately half ofthe patients with severe eosinophilic asthmain the Does Ranging Efficacy And safetywith Mepolizumab in severe asthma(DREAM) study were nonallergic,suggesting that besides the classical Th2pathway other immunologic mechanisms,such as the epithelial–innate lymphoid celltype 2 (ILC2) pathway, might be involvedin this asthma phenotype (12). We putforward the hypothesis that patientswith adult-onset (late-onset), nonallergicsevere eosinophilic asthma with repetitiveexacerbations are the best respondersto add-on treatment with anti–IL-5 oranti–IL-13 monoclonal antibodies. Ourhypothesis that innate lymphocytes suchas ILC2s and natural killer T cells driveeosinophilic airway inflammation insevere nonallergic asthma is based onthe following observations: (1) patientswith this severe asthma phenotype havefrequently chronic rhinosinusitis withnasal polyps as comorbidity, (2) ILC2sare strongly increased in eosinophilicnasal polyposis, and (3) ILC2s are ableto produce high amounts of IL-5 andIL-13 (22, 23). However, this hypothesisneeds to be tested appropriately by basic,translational, and clinical research inpatients with (nonallergic and allergic)severe asthma. If our hypothesis isconfirmed, the ILC2s pathway mightexplain the unusual pharmacology ofnonallergic severe asthma, which is poorlyresponding to topical treatment withICS but responds well to systemiccorticosteroids and the cytokine blockersanti–IL-5 and anti–IL-13.

Because the monoclonal antibodydupilumab targets the IL-4a receptor,which is a subunit common to the IL-4and IL-13 receptors, it has the abilityto interfere with both the Th2 pathway(including IL-4 signaling and IgE synthesis)as well as the ILC2 pathway. In an ICS-tapering RCT, dupilumab significantlyprevented mild asthma exacerbationscompared with placebo (24). However,at least two caveats should be underlinedbefore extrapolating these promising resultsto the treatment of patients with severeasthma in clinical practice. First, thepathogenesis of exacerbations elicited by

TRANSATLANTIC AIRWAY CONFERENCE

Brusselle and Bracke: Targeting Immune Pathways for Therapy in Asthma and COPD S323

reducing and stopping ICS and LABA inpatients with (partially) controlled asthmamight be fundamentally different fromnaturally occurring asthma exacerbationsin patients on persistent ICS1LABAtreatment. Second, because we hypothesizethat Th2 lymphocytes are very sensitiveto ICS, in contrast to ILC2s, the relativecontribution of these immunologicmechanisms/pathways might differaccording to whether the patient ison high-dose ICS or not.

In patients with neutrophilic asthma,add-on treatment with the macrolideazithromycin has been shown to decreasethe rate of exacerbations (25). In addition,a metaanalysis of RCTs of macrolidesin moderate to severe asthma hasdemonstrated that macrolides improvethe quality of life compared with placebo(26). These clinical data are in agreementwith the proven efficacy and safety ofmacrolides in other neutrophilic chronicairway diseases, such as cystic fibrosis,

non–cystic fibrosis bronchiectasis,diffuse panbronchiolitis, COPD, andbronchiolitis obliterans after lungtransplantation. However, the fact thatneutrophilic airway inflammation appearsto be a predictor of response to treatmentwith macrolides does not prove thatneutrophils are the main target in thesediseases. First, more specific antineutrophil-directed therapies such as brodalumab,a human anti–IL-17 receptor monoclonalantibody, have failed in patients withmoderate to severe asthma, although theinvestigators did not enrich the enrolledpatient population for the neutrophilicphenotype (27). Second, macrolideshave both immunomodulatory,antiinflammatory effects and antibioticeffects, encompassing the direct killingof microbes (e.g., Haemophilus influenzae),the prevention of biofilm formationthrough interfering with quorum sensing(e.g., Pseudomonas aeruginosa), and thestimulation of phagocytosis of microbes

by alveolar macrophages (28). Becausepopulation antimicrobial resistance dueto chronic treatment with macrolides is aconcern, the development of nonantibioticmacrolides is an attractive approach (29).It will be crucial to compare the effectsof the newer nonantibiotic macrolides insevere (neutrophilic) asthma not only withplacebo but also with the classical macrolideantibiotics, such as azithromycin. Ifazithromycin appears to be more efficaciousthan the newly developed nonantibioticmacrolide(s), this would suggest that thechronic bacterial colonization and infectionof the lower airways with microbes suchas H. influenzae and Haemophilusparainfluenzae is contributing to thepathogenesis of severe asthma.

COPD

The mainstay of current therapy for COPDis long-acting bronchodilators, including

allergen

epithelium

mast cell

lgE

TSLPTSLPP

TSLP

MHCII

TCR

IL-25

IL-33

IL-33

ILC2

IL-1

3

IL-4

, IL-1

3

goblet cell

pollutants, microbes, glycolipids

Th17 cellIL-17R

IL-17A

CXCL8GM-CSF

NKT cells

eosinophil

naive T-cell

B-cell

Th2 cell

IL-5

IL-13 IL-1

3

IL-13

IL-5

β2-adrenergic receptor

smooth muscle cell

ICS ± LABA

add-on treatment:

• anti-lgE (omalizumab)

• anti-lL-13

• anti-lL-4Rα (dupilumab)

ICS ± LABA

add-on treatment:

• anti-lL-5 (mepolizumab)

• anti-lL-13

• anti-lL-4Rα (dupilumab)

ICS ± LABA

add-on treatment:

• macrolides (azithromycin)

• anti-lL-17

• anti-lL-17R (brodalumab)

neutrophil

allergic eosinophilicasthma

nonallergic eosinophilicasthma

neutrophilicasthma

Figure 1. Simplified scheme of three different types of chronic airway inflammation in patients with asthma. In allergic eosinophilic asthma, Th2lymphocytes and mast cells drive eosinophilic airway inflammation in an allergen-specific, IgE-dependent manner. In nonallergic eosinophilic asthma,innate lymphocytes such as natural killer T cells (NKT cells) and innate lymphoid cells type 2 (ILC2) might contribute to airway eosinophilia via theproduction of IL-5. The mechanisms underlying neutrophilic asthma need to be elucidated, but the IL-17 pathway and CXCL8 have been associatedwith airway neutrophilia. GM-CSF = granulocyte/macrophage colony-stimulating factor; ICS = inhaled corticosteroids; LABA = long-acting b2-agonist;MHC = major histocompatibility complex; TCR = T cell receptor; TSLP = thymic stromal lymphopoietin.

TRANSATLANTIC AIRWAY CONFERENCE

S324 AnnalsATS Volume 11 Supplement 5| December 2014

LABAs and long-acting muscarinic receptorantagonists (30). Although LABAs andlong-acting muscarinic receptor antagonistshave additive effects on improvementof lung function and symptoms inpatients with COPD, they do not affectthe underlying pulmonary inflammation(Figure 2). Although a longitudinalstudy with bronchial biopsies hasdemonstrated that combination therapywith ICS and LABA significantly reducedthe number of CD81 T lymphocytes inthe bronchial mucosa of patients withCOPD, the antiinflammatory effects onother adaptive and innate immune cellnumbers were much less than seenin asthma (31). The chronic airwayinflammation in (ex)smokers withCOPD thus appears to be resistant totreatment with ICS (32).

The latest version of the GlobalInitiative for Chronic Obstructive LungDisease (GOLD) strategic documentrecommends adding ICS to long-actingbronchodilator therapy in patients withCOPD with frequent exacerbations(30). However, this recommendation

is an oversimplification. First, COPDexacerbations are heterogeneous in natureand etiology, encompassing exacerbationsthat are eosinophil-predominant, requiringtreatment with systemic corticosteroids,and exacerbations that are caused bybacterial lower respiratory tract infections,needing treatment with antibiotics (33).Second, the use of ICS in patientswith COPD has been associated witha significantly increased risk of pneumonia,including severe pneumonia and fatalpneumonia (34, 35). Therefore, it seemslogical to tailor the add-on therapy inthe frequent COPD exacerbator to thepredominant underlying phenotypeof exacerbations and concomitantcomorbidities. In patients with frequentexacerbations due to bacterial respiratoryinfections and/or bronchiectasis, ICSshould be avoided—or stopped—andmaintenance treatment with macrolidessuch as azithromycin during winter seasonis an effective alternative (36). In contrast,adding ICS to single or dual bronchodilatortherapy will be beneficial in patients withfrequent eosinophil-predominant COPD

exacerbations requiring repetitive bursts oforal corticosteroids.

The ultimate goal is to develop a safeand effective antiinflammatory treatmentfor COPD that has a disease-modifyingeffect (i.e., preventing the accelerated declinein lung function and reducing mortality).The community of respiratory researchersand clinicians can learn a lot from therheumatology field, where several disease-modifying antirheumatic drugs have beendeveloped in the past 2 decades. Theclassical example of disease-modifyingantirheumatic drugs is biologics targetingtumor necrosis factor (TNF)-a, such asthe monoclonal antibodies infliximab,adalimumab, or golimumab and the solubleTNF-a receptor etanercept (37). Treatmentof patients with rheumatoid arthritis withanti–TNFa biologics—most frequentlyin combination with methotrexate—hasrevolutionized the management of thisdisease, not only providing symptomaticbenefit but also and most importantlypreventing the relentless destructionof joints. Because TNF-a is elevated insputum and bronchoalveolar lavage of

epithelium

neutrophil

muscarinic receptor

BACTERIAL

COLONISATION Macrolides

• azithromycin

• erythromycin

Bronchodilators

• LABA

• LAMA

Anti-inflammatory treatment

• ICS

• anti-IL17, IL-17R antibodies

• anti-TNF antibodies

Anti-B cell antibodies

• anti-CD20 (rituximab)

• anti-CD22 (epratuzumab)

• anti-BAFF (belimumab)

B-cell

HEV

macrophage

Th17 cellTh1 cell

dendritic cell

CD8+ cytotoxic T-cell

INFLAMMATION

LYMPHOID

FOLLICLES

TLR TLR

NLRP3

procaspase-1

caspase-1

K+

efflux

pro IL-1βpro IL-18

IL-1βIL-18

K

NF-kB

INFLAMMASOME

Inflammasome targeting

• NF-kB inhibitors

• caspase-1 inhibitors

• IL-1β , IL-18 antagonists

• IL-1RI , IL-18R antagonists

IL-1βIL-18

IL-1RI

IL-18R

ATP

p2x

7

β2-adrenergic receptor

smooth muscle cells

ASC

Figure 2. Overview of pathogenic pathways and possible therapeutic targets in patients with chronic obstructive pulmonary disease. Please see textfor discussion. ASC = apoptosis-associated speck-like protein containing a caspase activation and recruitment domain; BAFF = B-cell–activating factorbelonging to the tumor necrosis factor family; HEV = high endothelial venules; ICS = inhaled corticosteroids; LABA = long-acting b2-agonist; LAMA: long-actingmuscarinic antagonist; NLRP = nucleotide-binding oligomerization domain-like receptor, pyrin domain-containing; NF = nuclear factor; TLR = Toll-like receptor.

TRANSATLANTIC AIRWAY CONFERENCE

Brusselle and Bracke: Targeting Immune Pathways for Therapy in Asthma and COPD S325

patients with COPD and increases furtherduring exacerbations, TNF-a blockers havebeen investigated in placebo-controlledRCTs in COPD. Treatment of patients withmoderate to severe COPD with the anti–TNF-a monoclonal antibody infliximabfor 24 weeks did not improve symptoms,lung function, exercise capacity, or therate of exacerbations (38). In contrast,infliximab treatment in COPD wasassociated with numerical increases inmalignancies, especially lung cancer, andpneumonias, implicating major safetyconcerns. Two lessons are to be learnedfrom this landmark study. First, TNF-aappears to play an important role in tumorimmunosurveillance in the respiratorytract. Smokers with COPD and/oremphysema have indeed a significantincreased risk of lung cancer comparedwith smokers without airflow limitationand without emphysematous destructionof the lung parenchyma (39). Second, incontrast to the sterile inflammation of thejoints in rheumatoid arthritis, the airwaysand lungs of smokers, and especiallypatients with COPD, are colonized withpathogenic bacteria, which might not onlycause acute exacerbations of COPD and/orpneumonias but also amplify the chronicpulmonary inflammation in stable COPD(40–42). Several studies using non–culture-based techniques (e.g., 16S ribosomal RNAsequencing) have demonstrated that theairway microbiome is altered in patientswith COPD compared with healthycontrol subjects, including an increase inProteobacteria such as H. influenzae andP. aeruginosa (43). It is worthwhile toinvestigate the efficacy and safety oftocilizumab and other anti–IL-6 therapiesin COPD, because tocilizumab has provento be efficacious in other chronic immunediseases such as rheumatoid arthritisand because IL-6 levels are significantlyincreased in patients with COPD, bothlocally in the lungs and systemically in theblood (in a COPD subgroup with persistentsystemic inflammation).

Chronic treatment with the macrolidesazithromycin and erythromycin has beenshown to significantly prolong the timeto the first exacerbation and reduce theexacerbation rate in patients with moderateto very severe COPD and a history ofrepeated exacerbations (36, 44). Cautionis warranted because patients with COPDare elderly people with multimorbidities(including cardiovascular disease) and

polypharmacy (including drugs thatcause QT prolongation), increasing therisk of hearing decrements and cardiacarrhythmias (45, 46). An additionalconcern is the increase in macrolide-resistant bacteria (and mycobacteria)within the COPD and general population(29), implicating that the developmentof nonantibiotic macrolides withantiinflammatory, immunomodulatoryeffects is much needed. Importantly, bycomparing the clinical efficacy of thenew nonantibiotic macrolides withazithromycin in patients with COPDwith frequent exacerbations, we will getnovel insights into the relative contributionof the antibiotic effects versus theantiinflammatory effects of azithromycin.These pivotal studies will offer us theopportunity to unravel the pathogenicrole of bacteria such as H. influenzae inCOPD and thus to test the vicious circlehypothesis of chronic infection andinflammation in COPD (40–42).

In severe COPD, increased numbersof lymphoid follicles have been observedaround the small airways and in thelung parenchyma (47–49). Theseperibronchiolar and intrapulmonarylymphoid follicles mainly contain B cellsand plasma cells and are local factoriesproducing antibodies in the mucosa(including immunoglobulin A, which issecreted into the airway lumen as secretoryIgA after transport through bronchialepithelial cells via the polymericimmunoglobulin receptor). These locallyproduced antibodies might be directedagainst viral and bacterial microorganismsin the respiratory tract of patients withCOPD and thus be protective (50).However, the lymphoid follicles might alsoproduce autoantibodies against epithelialantigens, posttranslationally modifiedproteins, or degraded extracellular matrixproteins. Several autoantibodies have beendetected in serum of a subgroup of patientswith (severe) COPD (51–53), althoughthe presence of elastin autoantibodies inCOPD is controversial (54, 55). Oxidativestress, an essential component in thepathogenesis of COPD, can induceincreased levels of highly reactive carbonylsin the lung, resulting in the formation ofimmunogenic carbonyl adducts on self-proteins. These carbonyl-modified proteinshave been shown to promote autoantibodyproduction (56). Because lymphoidfollicles and autoantibodies are supposed

to play a pathogenic role in this severeCOPD phenotype with autoimmuneemphysema, anti–B-cell–directed therapiesusing monoclonal antibodies against CD20(e.g., rituximab), CD22 (epratuzumab),IL-6 (tocilizumab), or BAFF (belimumab)merit investigation in well-designed RCTs(57). However, long-term studies willbe required to demonstrate efficacy onexacerbations and disease progression,and close monitoring of infectious adverseevents is warranted.

An interesting new avenue in thetreatment of chronic airway inflammationin COPD is targeting the inflammasomes,which are intracellular multiproteincomplexes in myeloid and epithelial cellsthat facilitate the activation of the cysteineprotease caspase-1 (58). Active caspase-1subsequently cleaves the inactive pro–IL-1band pro–IL-18 proteins into bioactive IL-1band IL-18, two major proinflammatorycytokines recruiting additional innateimmune cells and skewing adaptive Thelper cell responses toward Th1 and/orTh17 (Figure 2). Interfering with theinflammasome/caspase-1/IL-1b/IL-18pathway can be accomplished at severallevels: by inhibiting nuclear factor-kB–dependent up-regulation of pro–IL-1band pro–IL-18, by preventing the assemblyof the inflammasome complex or theenzymatic activity of caspase-1, and byblocking the interaction between secretedIL-1b/IL-18 and their membrane receptors.Because IL-1a, a danger signal that isindependent from the inflammasome/caspase-1 axis, is also increased in sputumand lungs of patients with COPD (59),blocking the common IL-1RI (whichbinds both IL-1a and IL-1b) might bemore efficacious than specific caspase-1inhibitors or IL-1b blockers (e.g., the anti–IL-1b monoclonal antibody canakinumab).However, deficient mucosal inflammatoryresponses to microbes may predisposepatients with COPD to respiratory tractinfections and pneumonia.

Predictors of TherapeuticResponse versusTherapeutic Targets

Finally, it is important to bear in mindthat a predictor of therapeutic response doesnot implicate that this predictor per seis a therapeutic target. The discordancebetween a predictor of response and

TRANSATLANTIC AIRWAY CONFERENCE

S326 AnnalsATS Volume 11 Supplement 5| December 2014

a therapeutic target is elegantly illustratedin the following examples. First, anincreased fraction of nitric oxide (NO)in the exhaled air of patients with asthma(either asthma-like symptoms such aschronic cough or a firm diagnosis ofasthma) is a predictor of a good responseto treatment with ICS (60). However,inhibiting inducible nitric oxide synthase(iNOS)—the enzyme responsible for theproduction of NO in the respiratorytract—did not improve lung function orsymptoms in patients with asthma (61).Second, the presence of neutrophilic

chronic airway inflammation is a predictorof therapeutic response to azithromycin(see above). However, specific antagonistsof mediators likely to be involved inthe chemotaxis and accumulation ofneutrophils into the airways have failed toshow clinical benefit in RCTs in COPD:antagonists of leukotriene B4, a CXCR2antagonist and a CXCL8 (formerly calledIL-8)-specific monoclonal antibody, wereineffective (32). Although we are eagerlyawaiting the results of clinical studiesinvestigating monoclonal antibodies againstIL-17, IL-17R, and IL-18 in COPD, the

current knowledge suggests that strategiesthat target the underlying causes of thepulmonary neutrophilia (e.g., smoking,oxidative stress, chronic infection, and soon) will be more successful than “cosmetic”strategies that merely decrease airwayneutrophilia without eradicating theunderlying drivers of neutrophilia. n

Author disclosures are available with the textof this article at www.atsjournals.org.

Acknowledgment: The author thanks LisaDupont for editorial assistance.

References

1 Barnes PJ. Immunology of asthma and chronic obstructive pulmonarydisease. Nat Rev Immunol 2008;8:183–192.

2 Wenzel SE. Asthma: defining of the persistent adult phenotypes.Lancet 2006;368:804–813.

3 Wenzel SE. Asthma phenotypes: the evolution from clinical tomolecular approaches. Nat Med 2012;18:716–725.

4 Moore WC, Meyers DA, Wenzel SE, Teague WG, Li H, Li X, D’Agostino RJr, Castro M, Curran-Everett D, Fitzpatrick AM, et al. National Heart L,Blood Institute’s Severe Asthma Research Program. Identificationof asthma phenotypes using cluster analysis in the Severe AsthmaResearch Program. Am J Respir Crit Care Med 2010;181:315–323.

5 Hogg JC, Timens W. The pathology of chronic obstructive pulmonarydisease. Annu Rev Pathol 2009;4:435–459.

6 Han MK, Agusti A, Calverley PM, Celli BR, Criner G, Curtis JL, FabbriLM, Goldin JG, Jones PW, Macnee W, et al. Chronic obstructivepulmonary disease phenotypes: the future of COPD. Am J Respir CritCare Med 2010;182:598–604.

7 Salvi SS, Barnes PJ. Chronic obstructive pulmonary disease in non-smokers. Lancet 2009;374:733–743.

8 Tashkin DP, Celli B, Senn S, Burkhart D, Kesten S, Menjoge S,Decramer M, Investigators US. A 4-year trial of tiotropium in chronicobstructive pulmonary disease. N Engl J Med 2008;359:1543–1554.

9 Holgate ST. Innate and adaptive immune responses in asthma. NatMed 2012;18:673–683.

10 Holmes AM, Solari R, Holgate ST. Animal models of asthma: value,limitations and opportunities for alternative approaches. Drug DiscovToday 2011;16:659–670.

11 Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, Keene ON, OrtegaH, Chanez P. Mepolizumab for severe eosinophilic asthma (DREAM):a multicentre, double-blind, placebo-controlled trial. Lancet 2012;380:651–659.

12 Brusselle GG, Maes T, Bracke KR. Eosinophils in the spotlight:eosinophilic airway inflammation in nonallergic asthma. Nat Med2013;19:977–979.

13 Djukanovic R, Wilson JW, Britten KM, Wilson SJ, Walls AF, Roche WR,Howarth PH, Holgate ST. Effect of an inhaled corticosteroid onairway inflammation and symptoms in asthma. Am Rev Respir Dis1992;145:669–674.

14 Laitinen LA, Laitinen A, Haahtela T. A comparative study of the effectsof an inhaled corticosteroid, budesonide, and a beta 2-agonist,terbutaline, on airway inflammation in newly diagnosed asthma:a randomized, double-blind, parallel-group controlled trial. J AllergyClin Immunol 1992;90:32–42.

15 Rabe KF, Vermeire PA, Soriano JB, Maier WC. Clinical management ofasthma in 1999: the asthma insights and reality in Europe (AIRE)study. Eur Respir J 2000;16:802–807.

16 Rabe KF, Adachi M, Lai CK, Soriano JB, Vermeire PA, Weiss KB,Weiss ST. Worldwide severity and control of asthma in children andadults: the global asthma insights and reality surveys. J Allergy ClinImmunol 2004;114:40–47.

17 Mehuys E, Van Bortel L, De Bolle L, Van Tongelen I, Annemans L,Remon JP, Brusselle G. Effectiveness of pharmacist intervention forasthma control improvement. Eur Respir J 2008;31:790–799.

18 Chung KF, Wenzel SE, Brozek JL, Bush A, Castro M, Sterk PJ, AdcockIM, Bateman ED, Bel EH, Bleecker ER, et al. International ERS/ATSguidelines on definition, evaluation and treatment of severeasthma. Eur Respir J 2014;43:343–373.

19 Humbert M, Beasley R, Ayres J, Slavin R, Hebert J, Bousquet J, BeehKM, Ramos S, Canonica GW, Hedgecock S, et al. Benefits ofomalizumab as add-on therapy in patients with severe persistentasthma who are inadequately controlled despite best availabletherapy (GINA 2002 step 4 treatment): INNOVATE. Allergy 2005;60:309–316.

20 Hanania NA, Wenzel S, Rosen K, Hsieh HJ, Mosesova S, Choy DF,Lal P, Arron JR, Harris JM, Busse W. Exploring the effects ofomalizumab in allergic asthma: an analysis of biomarkers in theextra study. Am J Respir Crit Care Med 2013;187:804–811.

21 Haldar P, Brightling CE, Hargadon B, Gupta S, Monteiro W, Sousa A,Marshall RP, Bradding P, Green RH, Wardlaw AJ, et al. Mepolizumaband exacerbations of refractory eosinophilic asthma. N Engl J Med2009;360:973–984.

22 Mjosberg JM, Trifari S, Crellin NK, Peters CP, van Drunen CM, Piet B,Fokkens WJ, Cupedo T, Spits H. Human IL-25- and IL-33-responsivetype 2 innate lymphoid cells are defined by expression of CRTH2and CD161. Nat Immunol 2011;12:1055–1062.

23 Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells–how did wemiss them? Nat Rev Immunol 2013;13:75–87.

24 Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, WangL, Kirkesseli S, Rocklin R, Bock B, et al. Dupilumab in persistentasthma with elevated eosinophil levels. N Engl J Med 2013;368:2455–2466.

25 Brusselle GG, Vanderstichele C, Jordens P, Deman R, Slabbynck H,Ringoet V, Verleden G, Demedts IK, Verhamme K, Delporte A, et al.Azithromycin for prevention of exacerbations in severe asthma(AZISAST): a multicentre randomised double-blind placebo-controlled trial. Thorax 2013;68:322–329.

26 Reiter J, Demirel N, Mendy A, Gasana J, Vieira ER, Colin AA, Quizon A,Forno E. Macrolides for the long-term management of asthma–a meta-analysis of randomized clinical trials. Allergy 2013;68:1040–1049.

27 Busse WW, Holgate S, Kerwin E, Chon Y, Feng J, Lin J, Lin SL.Randomized, double-blind, placebo-controlled study of brodalumab,a human anti-IL-17 receptor monoclonal antibody, in moderateto severe asthma. Am J Respir Crit Care Med 2013;188:1294–1302.

28 Brusselle GG, Joos G. Is there a role for macrolides in severe asthma?Curr Opin Pulm Med 2014;20:95–102.

29 Serisier DJ. Risks of population antimicrobial resistance associatedwith chronic macrolide use for inflammatory airway diseases. LancetRespir Med 2013;1:262–274.

30 Vestbo J, Hurd SS, Agusti AG, Jones PW, Vogelmeier C, Anzueto A,Barnes PJ, Fabbri LM, Martinez FJ, Nishimura M, et al. Globalstrategy for the diagnosis, management, and prevention of chronic

TRANSATLANTIC AIRWAY CONFERENCE

Brusselle and Bracke: Targeting Immune Pathways for Therapy in Asthma and COPD S327

obstructive pulmonary disease: GOLD executive summary. AmJ Respir Crit Care Med 2013;187:347–365.

31 Barnes NC, Qiu YS, Pavord ID, Parker D, Davis PA, Zhu J, Johnson M,Thomson NC, Jeffery PK, Group SCOS. Antiinflammatory effects ofsalmeterol/fluticasone propionate in chronic obstructive lungdisease. Am J Respir Crit Care Med 2006;173:736–743.

32 Barnes PJ. New anti-inflammatory targets for chronic obstructivepulmonary disease. Nat Rev Drug Discov 2013;12:543–559.

33 Bafadhel M, McKenna S, Terry S, Mistry V, Reid C, Haldar P,McCormick M, Haldar K, Kebadze T, Duvoix A, et al. Acuteexacerbations of chronic obstructive pulmonary disease:identification of biologic clusters and their biomarkers. Am JRespir Crit Care Med 2011;184:662–671.

34 Calverley PM, Anderson JA, Celli B, Ferguson GT, Jenkins C, JonesPW, Yates JC, Vestbo J; TORCH investigators. Salmeterol andfluticasone propionate and survival in chronic obstructive pulmonarydisease. N Engl J Med 2007;356:775–789.

35 Suissa S, Patenaude V, Lapi F, Ernst P. Inhaled corticosteroids in COPDand the risk of serious pneumonia. Thorax 2013;68:1029–1036.

36 Albert RK, Connett J, Bailey WC, Casaburi R, Cooper JA Jr, Criner GJ,Curtis JL, Dransfield MT, Han MK, Lazarus SC, et al. Azithromycin forprevention of exacerbations of COPD. N Engl J Med 2011;365:689–698.

37 Geiler J, Buch M, McDermott MF. Anti-TNF treatment in rheumatoidarthritis. Curr Pharm Des 2011;17:3141–3154.

38 Rennard SI, Fogarty C, Kelsen S, Long W, Ramsdell J, Allison J, MahlerD, Saadeh C, Siler T, Snell P, et al. The safety and efficacy ofinfliximab in moderate to severe chronic obstructive pulmonarydisease. Am J Respir Crit Care Med 2007;175:926–934.

39 Vermaelen K, Brusselle G. Exposing a deadly alliance: novel insightsinto the biological links between COPD and lung cancer. PulmPharmacol Ther 2013;26:544–554.

40 Sethi S, Evans N, Grant BJ, Murphy TF. New strains of bacteria andexacerbations of chronic obstructive pulmonary disease. N EnglJ Med 2002;347:465–471.

41 Alikhan MM, Lee FE. Understanding nontypeable Haemophilusinfluenzae and chronic obstructive pulmonary disease. Curr OpinPulm Med 2014;20:159–164.

42 Sethi S, Murphy TF. Infection in the pathogenesis and course of chronicobstructive pulmonary disease. N Engl J Med 2008;359:2355–2365.

43 Hilty M, Burke C, Pedro H, Cardenas P, Bush A, Bossley C, DaviesJ, Ervine A, Poulter L, Pachter L, et al. Disordered microbialcommunities in asthmatic airways. PLoS ONE 2010;5:e8578.

44 Seemungal TA, Wilkinson TM, Hurst JR, Perera WR, Sapsford RJ,Wedzicha JA. Long-term erythromycin therapy is associated withdecreased chronic obstructive pulmonary disease exacerbations.Am J Respir Crit Care Med 2008;178:1139–1147.

45 Wenzel RP, Fowler AA III, Edmond MB. Antibiotic prevention of acuteexacerbations of COPD. N Engl J Med 2012;367:340–347.

46 Ramos FL, Criner GJ. Use of long-term macrolide therapy in chronicobstructive pulmonary disease. Curr Opin Pulm Med 2014;20:153–158.

47 Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L,Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, et al. The natureof small-airway obstruction in chronic obstructive pulmonarydisease. N Engl J Med 2004;350:2645–2653.

48 van der Strate BW, Postma DS, Brandsma CA, Melgert BN, Luinge MA,Geerlings M, Hylkema MN, van den Berg A, Timens W, Kerstjens HA.Cigarette smoke-induced emphysema: a role for the B cell? Am JRespir Crit Care Med 2006;173:751–758.

49 Mori M, Andersson CK, Svedberg KA, Glader P, Bergqvist A,Shikhagaie M, Lofdahl CG, Erjefalt JS. Appearance of remodelledand dendritic cell-rich alveolar-lymphoid interfaces providesa structural basis for increased alveolar antigen uptake in chronicobstructive pulmonary disease. Thorax 2013;68:521–531.

50 Brusselle GG, Demoor T, Bracke KR, Brandsma CA, Timens W.Lymphoid follicles in (very) severe COPD: beneficial or harmful?Eur Respir J 2009;34:219–230.

51 Lee SH, Goswami S, Grudo A, Song LZ, Bandi V, Goodnight-White S,Green L, Hacken-Bitar J, Huh J, Bakaeen F, et al. Antielastinautoimmunity in tobacco smoking-induced emphysema. Nat Med2007;13:567–569.

52 Nunez B, Sauleda J, Anto JM, Julia MR, Orozco M, Monso E,Noguera A, Gomez FP, Garcia-Aymerich J, Agusti A. PAC-COPDInvestigators. Anti-tissue antibodies are related to lung function inchronic obstructive pulmonary disease. Am J Respir Crit Care Med2011;183:1025–1031.

53 Feghali-Bostwick CA, Gadgil AS, Otterbein LE, Pilewski JM, StonerMW, Csizmadia E, Zhang Y, Sciurba FC, Duncan SR. Autoantibodiesin patients with chronic obstructive pulmonary disease. Am J RespirCrit Care Med 2008;177:156–163.

54 Greene CM, Low TB, O’Neill SJ, McElvaney NG. Anti-proline-glycine-proline or antielastin autoantibodies are not evident in chronicinflammatory lung disease. Am J Respir Crit Care Med 2010;181:31–35.

55 Rinaldi M, Lehouck A, Heulens N, Lavend’homme R, Carlier V, Saint-Remy JM, Decramer M, Gayan-Ramirez G, Janssens W. AntielastinB-cell and T-cell immunity in patients with chronic obstructivepulmonary disease. Thorax 2012;67:694–700.

56 Kirkham PA, Caramori G, Casolari P, Papi AA, Edwards M, Shamji B,Triantaphyllopoulos K, Hussain F, Pinart M, Khan Y, et al. Oxidativestress–induced antibodies to carbonyl-modified protein correlatewith severity of chronic obstructive pulmonary disease. Am J RespirCrit Care Med 2011;184:796–802.

57 Chinen J, Shearer WT. Advances in basic and clinical immunology in2010. J Allergy Clin Immunol 2011;127:336–341.

58 Lamkanfi M, Dixit VM. Inflammasomes and their roles in health anddisease. Annu Rev Cell Dev Biol 2012;28:137–161.

59 Pauwels NS, Bracke KR, Dupont LL, Van Pottelberge GR, Provoost S,Vanden Berghe T, Vandenabeele P, Lambrecht BN, Joos GF,Brusselle GG. Role of IL-1a and the Nlrp3/caspase-1/IL-1beta axisin cigarette smoke-induced pulmonary inflammation and COPD.Eur Respir J 2011;38:1019–1028.

60 Smith AD, Cowan JO, Brassett KP, Filsell S, McLachlan C, Monti-Sheehan G, Peter Herbison G, Robin Taylor D. Exhaled nitric oxide:a predictor of steroid response. Am J Respir Crit Care Med 2005;172:453–459.

61 Singh D, Richards D, Knowles RG, Schwartz S, Woodcock A, LangleyS, O’Connor BJ. Selective inducible nitric oxide synthase inhibitionhas no effect on allergen challenge in asthma. Am J Respir Crit CareMed 2007;176:988–993.

TRANSATLANTIC AIRWAY CONFERENCE

S328 AnnalsATS Volume 11 Supplement 5| December 2014


Recommended