You are on page 1of 18

P­ri­me­r

­­­B­ro­nc­hi­ec­ta­si­s­­­­­­­­­­
James D. Chalmers1*, Anne B. Chang2,3, Sanjay H. Chotirmall4, Raja Dhar5 and
Pamela J. McShane6
Abstract | Bronchiectasis refers to abnormal dilatation of the bronchi. Airway dilatation
can lead to failure of mucus clearance and increased risk of infection. Pathophysiological
mechanisms of bronchiectasis include persistent bacterial infections, dysregulated immune
responses, impaired mucociliary clearance and airway obstruction. These mechanisms
can interact and self-​perpetuate, leading over time to impaired lung function. Patients
commonly present with productive cough and recurrent chest infections, and the diagnosis
of bronchiectasis is based on clinical symptoms and radiological findings. Bronchiectasis can
be the result of several different underlying disorders, and identifying the aetiology is crucial
to guide management. Treatment is directed at reducing the frequency of exacerbations,
improving quality of life and preventing disease progression. Although no therapy is licensed
for bronchiectasis by regulatory agencies, evidence supports the effectiveness of airway
clearance techniques, antibiotics and mucolytic agents, such as inhaled isotonic or hypertonic
saline, in some patients. Bronchiectasis is a disabling disease with an increasing prevalence and
can affect individuals of any age. A major challenge is the application of emerging phenotyping
and endotyping techniques to identify the patient populations who would most benefit from a
specific treatment, with the goal of better targeting existing and emerging treatments and
achieving better outcomes.

Bronchiectasis refers to abnormal, usually permanent, French physician René Laennec in 1819 (ref.4). Severe
dilatation of the bronchi (Fig. 1). It is a pathological or infections such as pneumonia and tuberculosis were
radiological term and a disease with its own diagnosis the predominant causes of bronchiectasis in the pre-​
code (J47.9) in the 10th revision of the International antibiotic era; thus, once antibiotics became available,
Statistical Classification of Diseases and Related Health bronchiectasis was expected to disappear as a clinical
Problems (ICD-10)1. Bronchiectasis as a disease is a clin- problem over time. Perhaps as a result of this assump-
ical syndrome that usually presents with cough, sputum tion, the development of drugs and clinical services
production and recurrent chest infections, along with for these patients received few investments, leading
other symptoms, such as malaise, chest discomfort, to bronchiectasis being described as an orphan or
haemoptysis (coughing of blood) and weight loss1. In neglected disease5.
terms of pathophysiology, bronchial dilatation leads to However, the predicted decline in bronchiectasis has
impaired mucociliary clearance, and failure to adequately not occurred, and the number of patients has risen inexor­
clear bacteria and mucus (the layer of gel-​like fluid that ably in the past decade, placing an increasing burden
facilitates the trapping of particulate matter and micro- on global health-​care systems6,7. As a result, there has
organisms that enter the airway) from the airways leads been a surge in interest in bronchiectasis8, with the gener­
to persistent infection, inflammation and further airway ation of international guidelines for treatment, the for-
damage2. Progressive airway damage can lead to impaired mation of international networks (including registries in
lung function, worsening symptoms and eventually res- the United States, Europe, India and Australasia), large-​
piratory failure and death3. Bronchiectasis can be the final scale randomized controlled trials and new studies into
common pathway of several infectious, inflammatory, mechanisms of disease9–11. Nevertheless, we are a long
allergic, genetic and degenerative disorders, and, there- way from fully understanding this disease or being able
fore, is the result of multiple pathophysiological pro- to offer the evidence-​based practice and personalized
cesses and one of the most complex and heterogeneous medicine that are now available in conditions such as
*e-​mail: jchalmers@
dundee.ac.uk syndromes in respiratory medicine. cystic fibrosis12.
https://doi.org/10.1038/ Bronchiectasis has been recognized as a major This Primer provides a comprehensive overview of
s41572-018-0042-3 clinical problem since its first description by the the progress that has been made in bronchiectasis, with

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 1

0123456789();
P­r i­m e­r

Author addresses Geographical differences. The overall prevalence of bron-


chiectasis varies geographically worldwide. Geographical
1
Scottish Centre for Respiratory Research, University of Dundee, Ninewells Hospital and variances may be due to several factors, including host
Medical School, Dundee, UK. factor susceptibilities, environmental exposures (for
2
Child Health Division, Menzies School of Health Research, Charles Darwin University, example, bronchiectasis is a common sequelae of tuber-
Darwin, Northern Territory, Australia.
culosis, and this fact is likely to be a major driver of
3
Department of Respiratory and Sleep Medicine, Queensland Children’s Hospital,
Children Centre for Health Research, Queensland University of Technology, Brisbane, differences in prevalences and patient characteristics
Queensland, Australia. among regions), local diagnostic practices and the nature
4
Translational Respiratory Research Laboratory, Lee Kong Chian School of Medicine, of available data sets24. In particular, in European and
Clinical Sciences Building, Nanyang Technological University, Singapore, Singapore. US populations, the variations in prevalence estimates
5
Department of Pulmonology, Fortis Hospital, Kolkata, India. may reflect differences in data capture between health-​
6
Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL, USA. care claims databases, primary-​care records and registries,
as much as true differences in prevalence between coun-
a focus on disease mechanisms and their implications tries. European countries report prevalence rates from
for patient management. 67 per 100,000 individuals in Germany to 362 per 100,000
individuals in Spain7,18,25. In 2013, the US prevalence was
Epidemiology estimated to be 139 per 100,000 individuals of >18 years
Prevalence and incidence of age25. In the same year, the UK prevalence was noted to
The ability to mine large health-​care databases for be 566.1 and 485.5 per 100,000 in adult women and men,
disease-​specific codes has provided increased insight respectively6. The highest prevalence of bronchiectasis
into epidemiological patterns in bronchiectasis. Data is reported in Asia, at 1,200 per 100,000 individuals of
indicate that bronchiectasis prevalence is increasing, and >40 years of age in China, although this rate is probably
this trend may be partially influenced by an increased a gross underestimate26. Accurate, reliable epidemiologi­
use of diagnostic CT scans13 and the establishment of cal data are lacking for large regions of the world, for
national bronchiectasis registries, both of which enable example, Eastern Europe, South America, the Middle
the identification of patients with bronchiectasis10,11. East and Africa.
Bronchiectasis can affect individuals of any age,
but the overall prevalence of bronchiectasis increases Sex differences. The reported ratio of affected males to
with age; the mean age of patients with bronchiecta- females is variable. In adults from the United Kingdom,
sis across continental Europe and Australia is 65 years Spain and the United States, women predominate
(range 59.4–68.3)14,15. In a US Medicare population (57–79% depending on the population studied)11,15,27. In
(which includes men and women of >65 years of age), Australia, 50.6% of patients are females but only 42% and
the annual percentage increase in the prevalence of 45% in China and South Korea, respectively14. Idiopathic
bronchiectasis was on average 8.74% from 2000 to 2007 bronchiectasis and non-​tuberculous mycobacteria
(ref.16). Across all age groups, the incidence of bronchi- (NTM) infection-​associated bronchiectasis are more
ectasis increased in the United Kingdom from 2004 to frequent in women than in men11,28,29, but the reasons
2013 (from 21.24 to 35.17 per 100,000 person-​years in for the increased susceptibility to bronchiectasis among
women and from 18.19 to 26.92 per 100,000 person-​ women are not well understood, although possible
years in men)6. As a corollary, bronchiectasis-​associated mechanisms have been reviewed30. In large parts of the
hospitalizations increased. The overall age-​adjusted world, smoking is more prevalent in men than in women
rate of all bronchiectasis-​associated hospitalizations and, as a result, bronchiectasis associated with smoking-​
in Germany increased at an average rate of 2.9% per related chronic obstructive pulmonary disease (COPD)
year from 2005 to 2011 (refs7,17). Similarly, in Spain, is more frequent in men than in women31. Smoking itself
an increasing incidence of bronchiectasis-​associated does not seem to be a risk factor for bronchiectasis, as
hospitalizations was observed from 2004 to 2013 (ref.18). the prevalence of radiological bronchiectasis in both
In children, bronchiectasis is commonly misdiag- male and female non-​smokers and smokers without
nosed as asthma19. In one study, 49% of children with COPD does not seem to be different32.
bronchiectasis on CT imaging had been diagnosed
with asthma, and the delay in receiving the correct Mortality
diagnosis of bronchiectasis was up to 14.8 years from Age-​adjusted mortality for patients with bronchiec­
symptom onset19. The prevalence of bronchiectasis is tasis is more than twice that of the general population6.
high in regions where children have early and recurrent Mortality correlates with the severity of disease but is not
respiratory infections20. For example, the prevalence strongly influenced by the underlying aetiology, with a
is estimated at 160 per 100,000 individuals in Māori/ few exceptions, such as rheumatoid arthritis and COPD,
Pacific Islander children in New Zealand and 1,470 which are associated with worse prognosis3,33–35.
per 100,000 individuals in Indigenous Australians of Predictors of mortality that are relevant to all possible
<15 years of age21,22. Furthermore, a disproportionate aetiologies include old age, low forced expiratory volume
number of US Alaskan native children in the YK Delta in 1 second (FEV1; the amount of air a person can force-
region is affected23. Whether the increased susceptibility fully exhale in 1 second in a forced expiratory volume
in these populations represents genetic differences or a test), low body mass index, previous hospitalizations and
more-​limited access to care, particularly for antibiotic frequent exacerbations (that is, unpredictable flare-​ups
treatment of childhood infections, is unclear. of symptoms often caused by infection)3,34–36

2 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

a Normal bronchus b Bronchiectasis Loss of cilia c


Smooth Increased
muscle cell mucus
Basement Destruction
membrane of wall
Goblet cell

Epithelial
Air cell
passageway
Cilia Normal
Mucus Cylindrical
Varicose
Blood vessel
Cystic
Wall

Fig. 1 | Morphology of healthy and dilatated bronchi. a | Structure of a healthy bronchus. b | Structure of a bronchus
with bronchiectasis. Note the airway dilatation, with obstruction of the airway with mucus, increased number and size
of mucous glands (goblet cells), bronchial wall damage and loss of cilia. c | Different morphologies of bronchiectasis.
Cylindrical bronchiectasis is the mildest form, whereas cystic dilatation is the most severe form, in which the morphology
of the bronchus is lost entirely.

Geographical variations of aetiologies Mechanisms/pathophysiology


Most of the evidence guiding our treatment of bron- The pathogenesis of bronchiectasis is complex, poorly
chiectasis has been generated in European adults, and understood and is likely to vary depending on the
very little data have been generated from developing underlying aetiology and important modifying factors.
countries or in children. The burden of bronchiectasis The airway epithelium represents a physical barrier built
seems to be substantial in countries and populations to resist infection through tight cellular junctions, mucin
with a high incidence of tuberculosis and other respir­ release, efficient ciliary function, antimicrobial peptide
atory infections24 (Fig. 2). There are also clear ethnic and production and active ion transport. All these mech-
cultural influences on the prevalence and presentation of anisms are thought to be impaired in bronchiectasis.
bronchiectasis, as demonstrated by the high incidence It has been postulated that there are two distinct phases
of childhood bronchiectasis in indigenous populations of disease: the first insult to the airway epithelium causes
in Australia22 or the high frequency of Pseudomonas initial bronchial dilatation and mucociliary impair-
aeruginosa infections reported in studies from China, ment2. The disease is hypothesized to be reversible at this
which may reflect different genetic susceptibilities to stage, if the original stimulus is controlled or removed42.
infection or different patterns of antibiotic use37. Genetics However, this initial phase predisposes to infection,
are likely to influence what bacteria an individual is most inflammation and failure of microbial clearance through
susceptible to — for example, in patients with cystic dysregulated immunity (Fig. 3), and, in some individ­
fibrosis, Staphylococcus aureus infection is much more uals, these insults establish a self-​perpetuating cycle that
common than would be expected by chance38. Some results in disease progression1,2.
organisms (such as Burkholderia cepacia) are very rare in Most evidence supports this disease model, but
bronchiectasis that is not due to cystic fibrosis39. Genetic important gaps exist in our understanding. For instance,
susceptibility to bronchiectasis development varies with inflammation and bronchiectasis may occur in the
ethnic origin: for example, cystic fibrosis is very rare in absence of infection43.
Asia compared with Western populations, and higher
frequencies of some primary ciliary dyskinesia (PCD)- Microbial colonization and infection
causing mutations have been reported in South Asian The airway in bronchiectasis is predisposed to microbial
individuals40. As a result of these variations, when using colonization due to mucus inspissation (thickening) and
clinical evidence to guide treatment, it is important to be impaired mucociliary clearance; these features, together
mindful of the patient population in which it has been with dysregulated immunity, may render commensal
generated, to ensure that the findings are applied to the organisms pathogenic under appropriate circumstances.
appropriate patients. Pulmonary infection is a major driver of airway inflam-
International registries should help to unravel some mation and bronchiectasis35,44,45 (Fig. 3). Airway damage
of these issues of patient representation. The European from severe or recurrent childhood lung infection may
Multicentre Bronchiectasis Audit and Research contribute to the development of bronchiectasis in later
Collaboration (EMBARC) is currently enrolling and has life, and protracted bacterial bronchitis (chronic, persis-
enrolled 12,000 European adult patients at the time of tent bacterial infection of conducting airways character-
writing, including patients from underrepresented popu­ ized by the presence of cough for >4 weeks) is implicated
lations in eastern and southern European countries10,41. in disease pathogenesis46,47. Bacteria, mycobacteria,
Registries now exist in Europe, the United States, Australia viruses and fungi have all been proposed to promote
and most recently in India, and more are expected to be the initiation and perpetuation of bronchiectasis, and
formed in additional countries in the coming years. bacterial load, rather than the specific pathogen, is the

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 3

0123456789();
P­r i­m e­r

100 worsening pathology 11,29,59. Mycobacterium avium


Children
90 complex is the most frequently isolated NTM in bron-
High-income countries
80 chiectasis, and patients with NTM infection are more
Low-income countries
70
likely than patients without NTM to develop Aspergillus
Adults spp.-associated disease, potentially indicating a shared
60
Cohort (%)

Europe and North America susceptibility factor60. Specific roles for viruses and
50 Africa, Asia and South America fungi are emerging but the mechanisms are currently
40 Australasia unclear61,62.
30 Collectively, infective agents contribute to the patho-
20 genesis of bronchiectasis by promoting airway dam-
age and increasing inflammation, and, by evading host
10
immune responses, pathogens can persist in the airway
0
and eventually become resistant to antimicrobial therapy63.
cy
c

us a

io al

PD

PA

α sea ive

ie sin
i

m
tio
PC
th

at nit
en

ns

n e
AB

y
fic yp
di ct
CO

th

nc
io

s
pa

ra

rm e
ci

e e

r
As
Dysregulated immunity
ct

f o ng
pi

de tit
su nn
io

efi
fe

As
Id

al o

tis Co
od

1 -A
- in

m C

Both innate and adaptive immune components are dys-


un
st
Po

regulated in bronchiectasis, and understanding their role


Im

Aetiology
is challenging because, paradoxically, both immunodefi-
Fig. 2 | Aetiologies of bronchiectasis grouped by regions and populations. Data ciency and immune hyper-​reactivity can lead to disease.
summarized from systematic reviews and publications in adults10,11,36,87,108,114,134 and
The paradoxical consequence of both hypo-​immune and
children110. However, one paper11 described some of these underlying aetiologies as
comorbidities (for example, primary immunodeficiency and inflammatory bowel
hyper-​immune function resulting in bronchiectasis sug-
disease); thus, the prevalence data may not be accurate. aUnclear whether these figures gests that a single defect or mechanism is improbable,
include post-​tuberculosis bronchiectasis. ABPA , allergic bronchopulmonary aspergillosis; and the extent of dysregulation also probably differs
COPD, chronic obstructive pulmonary disease; PCD, primary ciliary dyskinesia. among patients with bronchiectasis63–66. Inflammation
and destruction of the small airways can lead to bron-
chiectasis. This is evidenced by data from patients with
major determinant of the inflammatory response44. common variable immunodeficiency, in whom radio-
Most studies to date use traditional culture methods to logical signs (expiratory air trapping or mosaic attenua-
identify the causative bacterial pathogen, but molecu- tion) suggestive of inflammation-​associated alterations
lar approaches to characterize the microbiota have also in the small airways appear before bronchiectasis is
been performed in bronchiectasis, identifying organisms ­diagnosed and are potentially reversible67.
that are present in low abundance or difficult to detect
by routine culture48,49. The bacteria most frequently iso- Excessive immune response. In addition to providing
lated from airway secretions of patients with bronchi- a mechanical barrier against invading organisms, the
ectasis include Haemophilus influenzae, P. aeruginosa, epithelial cells recruit neutrophils by releasing pro-​
Moraxella catarrhalis, Streptococcus pneumoniae and inflammatory signals and increasing the expression of
S. aureus36,44,50. Virulence strategies vary between organ- surface glycoproteins that regulate phagocyte adhesion
isms and include structural-​mediated damage to the host and transendothelial migration63 (Fig. 3). These host
and inflammatory damage. Pathogens can also evade the responses become exaggerated in bronchiectasis, owing
host immune response via several mechanisms, such as to overwhelming persistent infections that perpetuate the
forming a biofilm, which acts as a shield, and undergo- pathogenic mechanisms68,69. In addition to airway inflam-
ing genetic changes (for example, genetic mutations can mation, there is evidence of systemic inflammation asso-
confer antimicrobial resistance). Some organisms, such ciated with increased levels of fibrinogen, a coagulation
as H. influenzae, can invade the airway epithelium and factor, which has been proposed as an indirect marker of
survive intracellularly, thereby circumventing or dysregu- disease severity70. The interaction between coagulation
lating the host immune response51,52. H. influenzae is one and inflammation is currently an area of intense study and
of the most frequent causes of chronic airway infections there is evidence that other systemic inflammatory
in patients with bronchiectasis, but the mechanisms are pathways are activated, including increased circulating
poorly characterized3,53,54. To date, such studies of the adhesion molecules and endothelial dysfunction, the
lung microbiome using sequencing technologies in bron- latter a marker of cardiac risk64,70.
chiectasis have confirmed a key role for Pseudomonas
and Haemophilus genera55,56. Bacteria can evade antibody Immunodeficiency. Immunodeficiencies, primary or
responses through cleavage or destruction of immuno- acquired, are associated with bronchiectasis, and fail-
globulins such as IgA57, whereas P. aeru­ginosa may utilize ure of adequate vaccine responses to S. pneumoniae
host IgG to block the activation of complement as a or H. influenzae have been described in some patients
virulence strategy58. P. aeruginosa is a key bronchiectasis-​ with bronchiectasis, indicating that their immune sys-
associated pathogen, associated with poorer clinical out- tems are unable to make coordinated B cell and T cell
come and higher mortality than those associated with responses to specific antigens71. Bronchiectasis is preva-
other pathogens35,45. lent in patients with chronic lymphocytic leukaemia and
NTM are intracellular pathogens that can poten- transporter associated with antigen presentation defi-
tially cause bronchiectasis directly or infect patients ciency syndrome72. In transporter associated with anti-
with established bronchiectasis, contributing to gen presentation deficiency syndrome, the expression

4 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

of major histocompatibility complex class I molecules eosinophils are usually associated with environmental or
is reduced, resulting in decreased antigen recognition allergic triggers and promote allergic TH2 cell-​mediated
by cytotoxic CD8+ T cells, impaired immune response responses. Airway neutrophilia is observed even in stable
and subsequent microbial persistence. Thus, cytotoxic bronchiectasis, but increases during infection and exacer­
T cells may play a key part in the development and pro- bations69,75,76 and contributes to disease pathogenesis
gression of bronchiectasis. Other congenital or acquired (Fig. 3). Neutrophils are recruited to the airway by chemo­
immunodeficiency states, such as HIV infection, are also attractants77, and cellular migration is regulated by a
associated with the development of bronchiectasis73. series of inter-​related and organized processes, including
altered surface receptor expression, increased cellular sig-
Cellular innate immunity. Airway neutrophilia is a hall- nalling and cytoskeletal manipulation in neutrophils, the
mark of bronchiectasis, although up to one-​quarter of endothelium and extracellular matrix78,79. Once neutro-
patients have eosinophil-​dominant airway inflamma- phils reach the airway, pathogens and the high concen-
tion74,75. The different cell populations may reflect differ- tration of inflammatory mediators trigger phagocytosis,
ent disease triggers, as neutrophils are mainly recruited degranulation and bacterial killing63. However, in bron-
by an infectious trigger and skew T cell responses towards chiectasis, despite their abundance, neutrophils fail at
T helper 1 (TH1) cell-​mediated inflammation, whereas removing bacteria, largely because phagocytosis is hin-
dered through several mechanisms, including cleavage
of receptors by neutrophil elastase and secreted defensins
NE NE NE (primarily released from neutrophils) and dysfunctional
Efferocytosis
intracellular signalling induced by the inflammatory
Macrophage CD35
IgG mileu80–83. Neutrophil elastase also directly cleaves com-
C3b/C4b CD11b/CD18 FcγR plement components bound to pathogens, preventing
• IL-1β • Cathepsin G opsonization and complement activation81. Neutrophil
Pathogen
CD88 • IL-8 • MMPs elastase is normally inhibited by natural anti-​proteinases,
P. aeruginosa • LTB4 • NE
• Proteinase 3
such as α1-antitrypsin and secretory leukocyte proteinase
H. influenzae • TNF
IL-17 • ROS inhibitor, but in the bronchiectasis airway, extensive neu-
NTM VCAM1 trophil recruitment and degranulation cause an excess of
Phagocytosed
C5a ICAM1 pathogen neutrophil elastase, and anti-​elastase defences are hin-
Biofilm
dered by multiple mechanisms, including cleavage and
Mucus TH17 cell CD8+ T cell
inactivation of secretory leukocyte proteinase inhibi-
tor76,84. This imbalance has been reported to damage air-
way epithelia, slow ciliary beat frequency and promote
Blood mucus production76,85–87. Neutrophil elastase levels cor-
Selectin
Goblet cell
vessel relate with disease severity, lung function, radiological
findings and sputum volume and have been evaluated
PSGL-1 Epithelial cell
• IL-8 as a potential inflammatory and prognostic marker in
• MIP-2 Neutrophil Endothelial cell
• TNF stable and exacerbating bronchiectasis that responds to
antibiotic therapy44,76,87.
Fig. 3 | Microbial infection and dysfunctional immunity contribute to the Similar to neutrophils, macrophage numbers are
pathophysiology of bronchiectasis. The respiratory epithelium recruits neutrophils by increased in bronchiectasis airways86,87. Patients with
releasing pro-​inflammatory signals (IL-8, macrophage inflammatory protein 2 (MIP-2; productive cough (with mucus production) seem to have
also known as CXC-​chemokine ligand 2 (CXCL2)) and tumour necrosis factor (TNF)) an increased burden of macrophages and macrophage-​
and increasing cell surface expression of intercellular adhesion molecule 1 (ICAM1) and derived neutrophil chemoattractants88,89. Macrophage
vascular cell adhesion protein 1 (VCAM1). C5a (from circulating complement) stimulates efferocytosis (the process in which apoptotic cells, particu-
airway macrophages to release TNF, IL-8, IL-1β and leukotriene B4 (LTB4), which promote
larly neutrophils, are cleared from the airway) is impaired
neutrophil recruitment77. Expression of CD11a/CD18 integrins on neutrophils (not
shown) and increased expression of selectin on endothelial cells promote neutrophil
in children with bronchiectasis and protracted bacterial
transmigration to the airway , where degranulation occurs. Key mediators of airway bronchitis, a forerunner of bronchiectasis90. Failure of
inflammation and destruction in bronchiectasis include reactive oxygen species (ROS), neutrophil clearance results in increased inflammatory
neutrophil elastase (NE), matrix metalloproteinases (MMPs), proteinase 3 and damage through unopposed granule product release and
cathepsin G, all of which are released by neutrophils56,76. Cleavage of neutrophil surface secondary necrosis of apoptotic cells. Neutrophil elastase
complement (CD35 and CD11b/CD18) and Fcγ receptors (FcγR; CD16 and CD32) by may impair neutrophil clearance by cleaving phosphati-
neutrophil elastase impairs opsonization and the phagocytic ability of neutrophils. The dylserine receptor from the surface of macrophages,
recognition of opsonized immunoglobulin G (IgG) targets and C3b/C4b and iC3b (not which is required for recognition of apoptotic cells91.
shown) complement components on microorganisms is also further compromised63.
Macrophages normally clear neutrophils by efferocytosis, a process impaired by
Impaired mucociliary clearance
neutrophil elastase release from excessive neutrophil degranulation in bronchiectasis.
Other airway immune cells, including lymphocytes, specifically cytotoxic T cells and
Mucociliary clearance is an important physiological
T helper 17 (TH17) cells, have a role in the pathogenesis of bronchiectasis by, respectively, self-​clearing mechanism for the airway. The epithelial
downregulating major histocompatibility complex class I molecules (thereby enabling cells lining the airways have hair-​like structures (cilia)
microbial persistence) and releasing IL-17 , an additional neutrophil chemoattractant207. on the surface; cilia beat in a tightly regulated manner
Ineffective microbial killing, persistent infection and inflammation coupled to to propel material towards the pharynx. Together with
mucus hypersecretion and airway epithelial damage all promote bronchiectasis. overlying mucus, cilia form the mucociliary escalator,
NTM, non-tuberculous mycobacteria; PSGL-1, P-​selectin glycoprotein ligand 1. which ensures that foreign agents entering the airway are

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 5

0123456789();
P­r i­m e­r

transported and either swallowed or expelled by cough- inflammation. Measurement of transepithelial ion
ing92. Damage to cilia and the associated mucociliary transport across the nasal epithelium (nasal potential
escalator can lead to bronchiectasis. Mucus trapping difference measurement) and CFTR mutation frequency
causes airway obstruction and dilatation and provides analyses in non-​cystic fibrosis-​associated bronchiectasis
a suitable environment for infection (Fig. 4). have suggested, in some cohorts, a higher than expected
Multiple factors may cause reduced ciliary beating, frequency of CFTR dysfunction103.
including cyanide produced by P. aeruginosa and neu-
trophil proteases, and deletion of airway cilia results in Other mechanisms
non-​inflammatory bronchiectasis and airway hyper-​ Oxidative stress (structural and functional alterations
reactivity93–95. Structural airway defects can contribute induced by reactive oxygen species) and hypoxia in
to impair the function of the mucociliary escalator and, the airway are induced by consumption of nutrients by
therefore, airway clearance. Anatomical defects of the inflammatory cells and bacteria and by reduced supply of
airway, such as tracheomalacia (softening of cartilage oxygenated blood to damaged lung segments. Oxidative
that maintains patency of the trachea), bronchomalacia stress is associated with airway damage in bronchiec-
(softening of cartilage in the bronchial walls) and tracheo­ tasis104,105. Activated immune cells in bronchiectasis are
bronchomegaly (a rare congenital disorder charac­ a rich source of reactive oxygen species, and increased
terized by a widening of the upper airways), can lead H2O2 levels in exhaled breath correlate with neutro-
to bronchiectasis through deficient airway clearance, phil burden, disease severity and lung function104,105.
excess secretions and recurrent infection96,97. Microbial infection further contributes to oxidative
Genetic conditions affecting cilia and their function stress and inflammation through phagocyte recruitment,
are also implicated in bronchiectasis. PCD is an auto- lipid peroxidation and the release of the stress response
somal recessive disease leading to dysfunctional cilia protein haem-​oxygenase-1 (ref.106). Vitamin D deficiency
and impaired mucociliary function54,98,99. In autosomal could also contribute to bronchiectasis pathogenesis;
dominant polycystic kidney disease, renal cysts develop however, it is unknown whether vitamin D deficiency,
because of defective cilia and ciliary polycystin-1 pro- by leading to impaired innate immunity, plays a part in
tein in renal epithelia100. Cilia of the airway epithelia also bronchiectasis or whether it is a result of bronchiectasis,
express polycystin-1, and radiology from patients with which can lead to a lack of outdoor sunlight exposure
autosomal dominant polycystic kidney disease reveals (owing to more-​severe disease and reduced mobility and
that substantial numbers of these patients have bronchi- exercise capacity) and, therefore, vitamin D deficiency107.
ectasis100. CFTR variants that result in the expression of
dysfunctional cystic fibrosis transmembrane conduct- Diagnosis, screening and prevention
ance regulator (CFTR) are also proposed to influence Diagnosis
the pathogenesis of bronchiectasis, even in the absence Diagnosing bronchiectasis requires clinical awareness of
of homozygosity for CFTR variants that would result in a this disease, to prescribe the necessary tests to confirm
diagnosis of cystic fibrosis101,102. Heterozygosity for these or rule out the diagnosis and detect possible underlying
CFTR variants may have a role in the development of diseases and comorbidities. Early diagnosis enables
diffuse bronchiectasis, at least in a subset of patients101,103. earlier achievement of treatment objectives (improving
CFTR mutations can result in abnormal ion transport the quality of life (QOL) of patients and possibly halt-
in the airways, leading to airway dehydration and ing disease progression)36,108, but many patients with
bronchiectasis are diagnosed at an advanced stage.
Chronic wet or productive cough, recurrent respiratory
• Excessive mucus secretion Dysfunctional infections, persistent pneumonia or chest radiography
• Airway obstruction and dilatation
• Persistent infections
cilia changes, haemoptysis, digital clubbing and dyspnoea can
Oxidative stress occur in both adults and children and should prompt
Hypoxia diagnostic tests for bronchiectasis. The presence of other
symptoms depends on the underlying characteristics of
the aetiology and patient population.
Biofilm CFTR variants
Mucus
Medical history, symptoms and signs. In adults, the
main symptom of bronchiectasis is chronic cough
with mucopurulent sputum (up to 80% of individuals),
Blood and bronchiectasis should be suspected in individuals
Endothelial cell Epithelial cell Goblet cell vessel
with productive couch and severe COPD or asthma or
Fig. 4 | Airway obstruction, impaired mucociliary clearance and other mechanisms chronic fatigue109. As disease severity increases, other
promoting bronchiectasis. Excess mucus production and the viscosity of mucus in symptoms (for example, dyspnoea, haemoptysis, weight
bronchiectasis promote airway obstruction and, therefore, bronchial dilatation. lost and fatigue) may occur. The relative prevalence of
Dysfunctional cilia (due to genetic mutations or acquired structural damage) and the
these symptoms and signs varies among global cohorts,
associated impaired mucociliary escalator further contribute to mucus retention and
airway obstruction. Oxidative stress (predominantly from immune cells), hypoxia (from dependent on the locality, aetiology and severity of
infection and inflammatory airway damage) and dysfunctional CFTR variants promote bronchiectasis and comorbidities.
and may lead to the development of bronchiectasis. Together, these mechanisms favour The clinical findings of children with bronchiecta-
persistent airway infection, inflammation and bronchial damage, thereby perpetuating sis also vary among global cohorts. Chronic cough is
the pathogenetic mechanisms underlying bronchiectasis. the most consistent symptom (35–100% of patients)

6 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

At any point in the lung, the internal luminal diameter


of the bronchus should be smaller than the external diam-
eter of the adjacent blood vessel. A bronchoarterial ratio
(BAR) of >1 is pathological. Lack of tapering of the bron-
chi and the presence of bronchi visible within 1 cm of the
pleura are additional radiological signs indicative of
bronchiectasis (Fig. 5).
However, the diagnostic criteria of bronchiectasis are
not univocal. Most studies define bronchiectasis if BAR
is >1 or >1.5, but using a single ratio irrespective of age
and the lung segment affected is, in the authors’ opinion,
problematic. Pathology-​based data and older studies
found a consistent BAR irrespective of lung inflation
and position, but studies using better CT technologies
reported that this ratio is age dependent and segment
dependent117–121. A study on 85 adults described a sig-
nificant positive correlation between age and BAR, and
41% of those of >65 years of age had a BAR of >1 (ref.122).
Extrapolation of the regression line from this study sug-
gests that the cut-​off BAR for normality should be at
Fig. 5 | Radiological features of bronchiectasis. High-​resolution CT scan slice
(reconstructed from a multi-​detector CT scan) shows bronchiectasis in a 7-year-​old child 0.4–0.5 in infants, and one study in children of 0–5 years
who first presented to the respiratory service with chronic cough. Dilatated bronchi and of age confirmed this value120. Experts have suggested
the adjacent blood vessels are visible, with the typical signet ring shape: cylindrical that a BAR of ≥0.8 should be the cut-​off used to define
bronchiectasis is present in the right lower lobe (blue arrow), and varicose-​to-cystic abnormality in individuals of <18 years of age when the
bronchiectasis is present in the right middle lobe, left lingula (yellow arrow) and lower clinical symptoms suggest bronchiectasis123.
lobe. Non-​tapering of the bronchi is visible in the left lower lobe (black arrow), and visible Several factors, for example, the presence of asthma
bronchi adjacent to the mediastinal pleura or within the outer 1–2 cm of the lung fields and elevated altitude, may increase the BAR and, there-
are present in the left lingula and lower lobe (red arrow). The aetiology of bronchiectasis fore, generate false-​positive results. Furthermore, a study
in this child is primary aspiration.
in adults with COPD described that the increased BAR
was related to a reduced diameter of the blood vessel and
and cough may resolve after treatment110,111. Of note, as not an increased diameter of the bronchus119. Some have
young children usually do not expectorate, the cough argued that the outer diameter of the bronchus should
is wet rather than productive112. Predictors of having be used instead of the inner diameter when measuring
bronchiectasis in children include failure to thrive and the BAR, and most studies do not specify what diam-
chest wall deformity (present in 45% and 95% of cohorts, eter was used120,123. In addition, the radiological bron-
respectively), the presence of recurrent (>3 per year) chial dilatation is probably a gradual process, and the
protracted bacterial bronchitis46 and failure of the wet type of CT may affect the ability to detect bronchiectasis.
cough to resolve with oral antibiotics (adjusted odds Contiguous 1-mm slices (from multi-​detector CT scans)
ratio = 20.9; 95% CI 5.4–81.8)113. Symptoms of dys­ are superior to conventional HRCT images (1-mm slice
pnoea and haemoptysis are generally rare in paediatric every 10 mm)124,125, and not all studies specify what type
cohorts in high-​income countries, where bronchiectasis of CT scans was used.
is generally diagnosed earlier (and hence patients have Thus, the diagnosis of bronchiectasis should not rely
milder disease at diagnosis) than in low-​income coun- on radiology only. For example, mild cylindrical bron-
tries. Cohorts based in low-​income countries generally chiectasis can be observed on CT in the absence of clini­
reported a higher prevalence of wheeze (up to 66%), cal symptoms in up to 20% of individuals of >65 years
haemoptysis (up to 41%) and digital clubbing (up to of age32,122. We propose that, in addition to radiology,
41%) than cohorts from high-​income countries outside the diagnosis includes a clinical phenotype supported by
of indigenous settings46,109–112. airway neutrophilia. A univocal definition of bronchi-
ectasis as a disorder would be particularly important for
Radiology. If bronchiectasis is suspected, radiological consistency across studies and advancement of interven-
examination should be performed. The diagnosis of tions aimed at treating mild bronchiectasis to prevent
bronchiectasis is objectively confirmed with a high-​ disease progression.
resolution CT (HRCT), which is the gold-​standard
diagnostic tool36,108,114 (Fig. 5). Volumetric CT, which ena- Initial evaluation
bles closer examination of the whole lung (that is, more Once bronchiectasis has been diagnosed, on the basis
images are acquired), has improved sensitivity and inter- of radiological findings and clinical presentation, addi-
observer agreement for the diagnosis of bronchiectasis, tional examinations should be performed to assess the
compared with standard HRCT scans115. Guidelines possible underlying aetiology, the severity of the disease
currently recommend the use of HRCT to confirm the and the presence of comorbidities and treatable traits.
diagnosis in patients with clinical symptoms of bronchi- Spirometry (a test to determine lung function by meas-
ectasis, but arguably volumetric CT with HRCT should uring FEV1 and forced vital capacity, which is the total
be the gold standard116. amount of air exhaled during a FEV test) is useful in

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 7

0123456789();
P­r i­m e­r

adults and older children (usually of >4 years of age). adults36,108,116,134. Nevertheless, despite extensive investiga-
When bronchiectasis is mild (typically with few lobes tions, the cause cannot be found in a substantial propor-
affected or only a mildly elevated BAR), lung function is tion of patients, who are then diagnosed with idiopathic
often completely normal126, particularly in children and bronchiectasis. The European Respiratory Society (ERS)
young adults with bronchiectasis living in affluent set- and Thoracic Society of Australia and New Zealand
tings. The classical spirometry abnormality associated guidelines recommend a similar standard set of tests
with bronchiectasis is an obstructive profile in which for adults with bronchiectasis36. The Thoracic Society of
airway collapse obstructs expiratory airflow, resulting Australia and New Zealand guidelines includes children
in a reduced FEV1 but normal lung volumes; a mixed and recommends a sweat test for children to identify
obstructive–restrictive pattern may also be present, with cystic fibrosis. Using an algorithm improves the ability to
both FEV1 and forced vital capacity reduced3,127. diagnose an underlying condition that leads to a change
Obtaining a lower airway secretion specimen (usu- in management134, as shown in a study in which the use
ally sputum) is another necessary component of the of an algorithm reduced idiopathic bronchiectasis as the
initial evaluation in both adults and children, particu- attributed aetiology from 42% to 29%. The additional
larly to guide antibiotic therapy and identify NTM and tests to be prescribed should be guided by the medical
P. aeruginosa. history (past and present) and clinical findings.
Bronchoscopy is not required routinely in adults who
are able to spontaneously expectorate sputum for micro- Prognosis
biology assessment, but may be useful if foreign body There is a large variation in prognosis, with some
obstruction is suspected or to evaluate patients who are patients having mild bronchiectasis for many years and
unable to provide good-​quality sputum samples116. Some others progressing rapidly with frequent exacerbations
centres undertake bronchoscopy in children routinely, and accelerated lung function decline34,76. The reversi-
for lower airway assessment and to exclude foreign bod- bility of radiologically defined bronchiectasis (which is
ies; information from bronchoscopy helped to guide defined when the increased BAR has returned to normal
antibiotic or corticosteroid treatment in 41% of cases128. values) is possible when the cylindrical bronchiectasis
In two systematic analyses of bronchoscopic findings in is treated early (Fig. 6). This has been described in chil-
children129,130, five airway lesions were described, ranging dren but is almost certainly also true in adults. In one
from bronchitis to an obliterative lesion, and the extent study, bronchial dilatation resolved completely in 6 out
of disease on CT correlated to the type of airway lesion. of 21 children with radiologically defined bronchiecta-
sis when HRCT scans were repeated 2–43 months fol-
Severity scores lowing intensive medical therapy135. In addition, when
There are no specific disease severity scoring systems primary immunodeficiency disorders are diagnosed
for children with bronchiectasis, but prognostic and early and optimally managed, bronchiectasis is not a
severity scores have been developed in adults3,9,131. The consequence136, and if already present, bronchiectasis
two scores that have been most extensively studied are can improve with optimal treatment (Fig. 6).
the Bronchiectasis severity index (BSI) and the FACED Mortality risk in bronchiectasis can be thought of as
score3,131. The BSI was developed and validated in a pro- an interaction between age, lung damage, lung infection,
spective multicentre European study of >1,000 patients disease activity and comorbidities. Thus, assessment of
and was specifically designed to predict not only mortal- outcome should take into account the measurement of all
ity but also hospital admissions and future exacerbations of these components, in both adults and children. Lung
and to identify patients with worse QOL3,9,132. The BSI damage may be considered in terms of functional capacity
has been shown to be a robust and reproducible marker (which can be measured through the Medical Research
of disease severity and adverse outcomes in multiple Council Dyspnoea score or other measures of functional
studies across Europe, Asia and the United States3,9. impairment), the extent of disease on CT scan or lung
The FACED score is an acronym for FEV1, age, colo­ function testing. Disease activity is best evaluated by the
nization with P. aeruginosa, extension of radiologi­cal frequency of exacerbations and severe exacerbations.
bronchiectasis and dyspnoea. Although it contains Comorbidities may be causes of bronchiectasis (such as
similar variables to the BSI, it attributes a higher propor- rheumatoid arthritis) or unrelated diseases, such as car-
tion of points to age and does not include exacerbations. diac disease3,27,33,45,76. Thus, patients with frequent exacer-
The FACED score predicts mortality but has not bations, a high symptom burden, old age, more-​extensive
performed well as a predictor of morbidity in most radiological disease and chronic infection, particularly
validation studies9. with P. aeruginosa, are the patients with the most rapid
clinical decline. Among these risk factors, exacerbations
Evaluation of possible underlying diseases seem to be the most important modifiable risk34.
Upon confirming the presence of bronchiectasis, patients It is highly likely that factors other than the under­
should also be evaluated for possible underlying dis- lying aetiology and the discussed mortality risks also
eases3,15,27. Bronchiectasis is a heterogeneous disease affect outcomes; examples include access to health care,
and is associated with multiple underlying aetiologies, the efficacy of (and adherence to) interventions, the
coexisting diseases and overlapping syndromes15,27,33,36,133 severity of bronchiectasis when first diagnosed43 and
(Table 1) that vary among regions (Fig. 2). The search for patient and social factors137. This is clearly described in
the underlying diseases is clinically important, as the the paediatric literature, in which early diagnosis and
aetiology guides clinical management in children and appropriate interventions can alter the natural history.

8 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

Table 1 | Aetiologies associated with the development of bronchiectasisa


Key features Examples of specific aetiology Key diagnostic test(s)b
Post-infectious
• Largest attributable cause of bronchiectasis in some Tuberculosis Medical history
cohorts1,15,24. Of note, the bias of data recall limits the Pneumonia206 Medical history
validity of this aetiology
• Any severe past infection may lead to bronchiectasis
Immunodeficiency
• The link between immunodeficiency and bronchiectasis Primary: agammaglobulinaemia and hypoglobulinaemia, IgA , IgG and IgM levels
relates to increased susceptibility to pulmonary infections, causing recurrent deep infections
causing recurrent and/or persistent infection, inflammation Primary: selective hyperglobulinaemia (for example, Specific immunoglobulin
and immune dysregulation hyper IgE syndrome and hyper IgM syndrome), causing levels
• Immunodeficiencies are important to identify , as their recurrent deep infections
identification changes clinical management
• Thus, the standard set of baseline tests recommended by Primary: neutrophil abnormalities (function and/or In-​depth immune tests
the European Respiratory Society , the Thoracic Society of movement), causing chronic granulomas and recurrent
Australia and New Zealand and other guidelines includes infections
serum IgG, IgA , IgM and IgE levels Primary: other rare causes (for example, bare Genetic and in-​depth
• There is an increasingly wide array of primary innate, lymphocyte syndrome) immune tests
humoral, phagocytic and complement immune deficiency
and dysregulation identified only through in-​depth immune Secondary: secondary immunodeficiencies (for Serology and medical
tests aided by genetic confirmation208 example, HIV infection and chemotherapy-​induced history
immunosuppression)
Impaired mucociliary clearance
• Primary impaired mucociliary clearance is associated with Cystic fibrosis Sweat test and genotype
genetic diseases, including cystic fibrosis, PCD and Jeune PCD Nasal nitric oxide test, cilia
syndrome)92,209,210 biopsy and motility test, and
• Conditions associated with respiratory muscle weakness genetic analysis
and impaired cough effectiveness (inspiratory and
expiratory forces) Other less common conditions, for example, Various clinical appearances
asphyxiating chest wall abnormality , congenital muscle and genetic tests dependent
dystrophies and myopathies on the condition
Previous lung injury
Wide group of conditions, including: Primary aspiration (due to dysphagia that is often, but Swallowing assessment (for
• Aspiration (the entry of material (such as food, drink or not always, related to neurological abnormality) example, video fluoroscopy)
stomach contents) from the gastrointestinal tract into the Secondary aspiration (due to gastroesophageal reflux). Endoscopy and motility
larynx and lower respiratory tract) Of note, gastroesophageal reflux disease as a cause of testing
• Bronchiolitis obliterans (inflammation of the smallest bronchiectasis is controversial11
airways of the lungs leading to obstruction) Bronchiolitis obliterans (the end product of a large CT scan
number of respiratory conditions, including infections,
graft-​versus-host disease and autoimmune conditions)
Interstitial lung disease (drug induced or due to Medical history
occupational inhalation exposure)
Airway lesions
Structural abnormalities (primary , that is, congenital, Congenital tracheobronchomegaly , primary or Chest CT and bronchoscopy
or secondary , for example, due to injuries) of the airway secondary tracheal stenosis or cartilage abnormalities
can promote the pathophysiological mechanisms of Obstructive (for example, foreign body or external Medical history , chest CT
bronchiectasis compression) and bronchoscopy
Tracheobronchomalacia Bronchoscopy
Concurrent other chronic respiratory diseases
• Both chronic obstructive pulmonary disease (COPD) COPD Lung function and smoking
and bronchiectasis share common features (including history
spirometry abnormality and chronic cough) and aetiologies Asthma Lung function — tests for
(for example, childhood respiratory infections, inhalational airway reactivity
injury and tuberculosis)
• However, some controversy exists121 regarding the Allergic bronchopulmonary aspergillosis should be Elevated anti-​Aspergillus
causative role of COPD in bronchiectasis, as it remains diagnosed according to guidelines212 spp. IgE levels, total IgE
unknown whether one disease precedes another or a supported by raised IgG
previous misclassification occurred and/or Aspergillus spp.
• Adults with COPD and bronchiectasis have a poorer prognosis precipitin and eosinophil
than adults with bronchiectasis without co-​existing COPD32,33 assessment
• Asthma can be a comorbidity , but whether it causes
bronchiectasis is controversial
• One study described that 40% of newly referred adults
with ‘difficult asthma’ (and who had a CT scan) were
found to have bronchiectasis, suggesting that disease
misclassification exists211

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 9

0123456789();
P­r i­m e­r

Table 1 (cont.) | Aetiologies associated with the development of bronchiectasisa


Key features Examples of specific aetiology Key diagnostic test(s)b
Connective tissue disease and/or autoimmune
• Important causes in some cohorts, predominantly in Inflammatory bowel disease Colonoscopy
North America (28% of cohort)213 Rheumatoid arthritis Serum autoantibody
• Connective tissue diseases overlap with immunodeficiency assessment
syndromes208
• A wide array of connective tissue diseases can lead to Systemic lupus erythematosus Serum autoantibody
bronchiectasis through the development of interstitial lung assessment
disease, the use of immunosuppressants or other unknown Sjögren syndrome Serum autoantibody
mechanisms assessment
• Bronchiectasis can precede the development of other
features of connective tissue disease
• Inflammatory process in the lungs can provoke
autoimmune responses, such as the generation of
citrullinated peptide antibodies against antigens released
from activated neutrophils
Other syndromes
Bronchiectasis can be associated with conditions that are not Ataxia-​telangiectasia α-​Fetoprotein and genetic
predominantly pulmonary analysis (ATM gene)
Marfan syndrome Clinical characteristics
(defined set exits) with or
without the presence of
fibrillin-1 (FBN1) mutation
Polycystic kidney disease Renal ultrasonography and
genetic analysis
Velocardiofacial syndrome Genetic analysis (22q11
deletion)
Yellow nail syndrome Clinical characteristics. In
some patients, mutations
in FOXC2 may be found
Others
• Being born premature is associated with bronchiectasis in Prematurity Medical history
children and adults200. In a small cohort of adult preterm α1-Antitrypsin deficiency Levels and function of
survivors, 3 of the 46 (6.5%) adults had CT-​defined α1-antitrypsin
bronchiectasis 200

• α1-Antitrypsin deficiency is associated with bronchiectasis


with emphysema; however, guidelines from Europe, Australia
and New Zealand and the United Kingdom do not advocate
screening for it in the absence of emphysema36,108,116
Ig, immunoglobulin; PCD, primary ciliary dyskinesia. aThe table is not a complete list of all possible aetiologies, as some rare diseases have been omitted for brevity.
b
Different guidelines may recommend different diagnostic tests (dependent on the setting and the prevalence of different conditions).

Screening and prevention trials, some believe that little can be done to prevent
As disease confirmation requires a CT scan (thus expo- the disease and its progression. Preventive strategies
sure to radiation), population screening is not advocated largely relate to preventing acute respiratory infections
when symptoms are absent. However, one Korean-​based (summarized in Box 1).
study that included screening for bronchiectasis as part
of a health-​screening programme reported that 9.1% of Management
the 1,409 adults (23–88 years of age) examined had radio­ The treatment of bronchiectasis aims at stopping the
logical bronchiectasis138. Although they were previously self-​p erpetuating pathogenetic mechanisms. This
undiagnosed, 54% of the individuals that were identi- approach includes strengthening the mucociliary esca-
fied to have bronchiectasis had respiratory symptoms, lator, redu­cing airway inflammation and eradicating
which may indicate that the bronchiectasis was clinically bacterial infections (if present)36. As bronchiectasis can
relevant, and 15% had a previous history of tuberculo- result from several specific conditions (Table 1), the first
sis. These results suggest that screening with CT could key step in treatment is identifying and treating the
identify unrecognized bronchiectasis in adults, and the underlying disorder, while also optimally managing
increasing use of CT for other screening programmes comorbid conditions.
(such as lung cancer screening) is likely to identify bron-
chiectasis in a high proportion of patients. No interna- Airway clearance methods
tional guidelines currently suggest population screening The rationale for promoting airway clearance is removing
for bronchiectasis. purulent secretions from the airways, which ameliorates
Since the 1940s, clinicians have recognized that bron- the inflammation and improves control of symptoms
chiectasis is reversible when treated early. Nevertheless, such as sputum production and mucus plugging92,139–141.
in the absence of evidence from randomized controlled Several studies have demonstrated the benefits of

10 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

a is another mucoactive agent that has been tested in bron-


chiectasis, but a study in 461 patients with bronchiectasis
concluded that inhaled mannitol did not reduce exacer-
bation frequency, although there were improvements in
time to first exacerbation and QOL146.
Although mucolytics such as N-​acetylcysteine and
carbocysteine are used widely in clinical practice, there is
no published evidence of benefit from these agents147,148.
International guidelines for bronchiectasis do not rec-
ommend use of recombinant DNase in adults or chil-
b
dren, because a multicentre randomized controlled trial
in adult patients with bronchiectasis and P. aeruginosa
infection showed no benefit and the possibility of harm
in terms of increased frequency of exacerbations and
more-​rapid decline in FEV1 (ref.149). Recombinant DNase
is effective in patients with cystic fibrosis, and reasons
for the discrepancy in the response to DNase between
patients with bronchiectasis and patients with cystic
fibrosis have been the subject of much debate but have
Fig. 6 | Resolution of bronchiectasis in a child. a | Two slices of the initial chest CT scan never been clearly explained. One study suggested that
of a 5-year-​old child who had had common variable immunodeficiency for >3 years and DNase treatment of sputum from patients with bron-
was receiving intravenous immunoglobulin every 4 weeks. The CT scan, undertaken at
chiectasis made the sputum more-​difficult to transport
the first consultation with a paediatric respiratory physician, shows areas of cylindrical
(blue arrow) and varicose (yellow arrow) bronchiectasis in the left lower lobe. b | The along the mucociliary escalator150.
follow-​up corresponding chest CT scan slices of the same child 8 years later. The CT was
taken as the child was clinically less stable (more exacerbations, although spirometry Low-​dose macrolide therapy
values were stable), but shows improvement from the initial scan with resolution of Macrolides are a class of orally administered antibiotics
cylindrical bronchiectasis and improvement of the varicose bronchiectasis changes that have both anti-​inflammatory and immunomodula-
(yellow arrows). When well, the child did not have a daily cough. tory properties and, therefore, are beneficial to patients
with chronic pulmonary diseases, such as diffuse pan
bronchiolitis, cystic fibrosis and bronchiectasis151,152.
clearance methods, which include physiotherapy tech- A series of pivotal studies have provided strong evidence
niques and inhaled agents (see below)142,143. Techniques (in both adults and children) in favour of prescribing
used worldwide can be self-​performed, require assistance low-​dose macrolide for the reduction of the frequency
of another person, such as a physiotherapist, or require of exacerbations in patients with bronchiectasis. In the
devices that generate positive expiratory pressure to BLESS randomized controlled trial, long-​term low-​
mobilize secretions. The ERS guidelines for bronchiec- dose erythromycin significantly reduced the frequency
tasis recommend daily physio­therapy techniques, such as of protocol-​defined pulmonary exacerbation and also
active cycle of breathing techniques, autogenic drainage reduced 24-hour sputum production and lung function
and oscillating posi­tive expiratory pressure, which are all decline153. In the EMBRACE study, azithromycin was
techniques that patients can conduct independently in associated with a significant 62% reduction in exacer-
their own homes36. For example, the active cycle technique bation rates compared with placebo, with no significant
includes breathing control (a period of relaxed breath- changes in pulmonary function154.
ing) and thoracic expansion exercises (deep breathing, Similar results were observed in the BAT study, in
which expands the alveoli and increases expiratory flow which azithromycin significantly reduced the time to
behind secretions) followed by huffing, which forces first exacerbation and the number of patients experienc-
the sputum into the larger airways for expectoration. The ing exacerbations, with improvements in QOL and lung
ERS guidelines also strongly advocate pulmonary rehabil- function also observed. However, gastrointestinal adverse
itation for adult patients, as it improves exercise tolerance effects were observed in 40% of individuals in the azithro-
and health-​related QOL (HRQOL) 144. Pulmonary mycin group, and the macrolide resistance rate was 88%
rehabilitation is not used in children. in sputum bacteria in the azithromycin group, compared
with 26% in the placebo group155. In a randomized con-
Adjuvant inhaled agents. Inhaled mucolytics (which loosen trolled trial in 89 Indigenous Australian children, azithro­
the mucus) and hyperosmolar agents (which incre­­ase the mycin reduced the exacerbation rate by half; however,
hydration of the airway) can facilitate mucus clearance similarly to the studies in adults, increased numbers of
and are adjuvant therapies for airway clearance, that children carrying nasopharyngeal bacteria resistant to
is, they are used alongside physiotherapy techniques. macrolides were observed with azithromycin treatment156.
Hypertonic saline hydrates airway mucus and increases These randomized controlled trials suggest that
ciliary beat frequency, thereby improving airway clear- macro­lides are beneficial in the treatment of bronchiecta-
ance143,145. A small trial showed that inhalation of hyper- sis for patients with frequent exacerbations in both adults
tonic saline or isotonic saline has a similar effect on risk and children. Important considerations in prescribing
of exacerbation, QOL and respiratory function tests over macrolides are that a substantial number of patients will
12 months143, although larger trials are needed. Mannitol develop gastrointestinal adverse effects, which are likely

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 11

0123456789();
P­r i­m e­r

to be dose related155. In addition, NTM infection should increases the rate of exacerbations44. Inhaled antibiotics
be excluded before macrolide treatment, to avoid the are only used in patients with confirmed bacterial air-
risk of inducing macrolide-​resistant NTM157. The pos- way infection and are often restricted to patients with
sibility of less common adverse effects, such as hearing P. aeruginosa infection158–162. Tobramycin, colistin and
loss and cardiovascular adverse effects, should also be gentamicin are the most commonly used in clinical prac-
considered152. The development of antibiotic resistance tice. However, randomized trials have given conflicting
in oropharyngeal or sputum bacteria is a concern, but data on the effectiveness of inhaled antibiotics. All of the
no data are yet available on whether macrolides lose effi- trials discussed below were conducted in adults.
cacy in patients with bronchiectasis with time as a result In a double-​blind randomized controlled study
of long-​term use. Studies with a follow-​up time >1 year on 74 patients with bronchiectasis and P. aeruginosa
would be required to verify this hypothesis. infection163, P. aeruginosa was eradicated in 35% of the
patients who received tobramycin solution for inhalation,
Inhaled antibiotics whereas there was no change in the placebo group. The
The main objective of using inhaled antibiotics in bron- adverse effects, such as cough, dyspnoea, wheezing and
chiectasis is the reduction of bacterial load, as high bac- non-​cardiac chest pain, were more frequent in patients
terial loads increase the local and systemic inflammatory receiving tobramycin solution for inhalation, but these
response, which worsens symptoms of the disease and symptoms did not limit therapy. Increased respiratory
symptoms with tobramycin have been reported in other
Box 1 | Major preventive measures for the development of bronchiectasis trials164,165. Approximately 10% of patients treated with
inhaled antibiotics seem to experience bronchospasm,
In utero strategies
with rates reported in trials of tobramycin or gentamicin
• Improving fetal health: improves in utero lung development, thereby preventing
higher than those reported for other drugs166.
intrauterine growth restriction, fetal intrathoracic space, intrauterine infection or
inflammation198; in utero alterations in lung structure and function can influence
In a randomized controlled study of colistin in
future respiratory outcomes198 patients with bronchiectasis and chronic P. aeruginosa
• Preventing tobacco smoke exposure: improves in utero lung development, alters
infection, the primary end point was not met, as the
immune development and programming, thereby reduces the risk of acute respiratory median time to first exacerbation was not significantly
infections (ARIs)199; infants born to mothers who smoked during pregnancy have different between the colistin and placebo groups
diminished lung function, altered respiratory chemoreception and increased risk of ARIs (165 days and 111 days, respectively)158. P. aeruginosa
• Preventing prematurity: prematurity is associated with increased ARIs, reduced lung density was reduced after 4 weeks and 12 weeks, and
function and increased risk of chronic obstructive pulmonary disease (COPD), and is a the QOL was improved after 26 weeks in the colis-
risk factor for the development of bronchiectasis200 tin group compared with those receiving placebo158.
Postnatal strategies Although the study failed to meet its primary end point,
colistin is widely used in Europe and worldwide for the
• Encouraging breastfeeding: passive transfer of immunoglobulins through breast milk
reduces ARIs; a case–control study found that breastfeeding was a protective factor control of chronic P. aeruginosa infection in patients
for bronchiectasis201 with bronchiectasis158.
• Preventing tobacco smoke exposure and active smoking: reduction of passive smoke
A randomized controlled trial of nebulized gen-
reduces the risk of ARIs, asthma, impaired lung development and the risk of COPD. tamicin in patients with bronchiectasis showed that,
Systematic reviews have shown increased risk of COPD in adults who smoke and after 1 year, there was reduced sputum bacterial density
increased risk of ARIs and reduced lung function in children exposed to smoke202 in the gentamicin arm, with a 30.8% eradication rate in
• Reducing exposure to pollution (for example, biomass combustion and other air patients with P. aeruginosa infection and 92.8% eradi-
pollution): reduces the risk of respiratory disorders, COPD, respiratory infections and cation rate in those with other pathogens. Thus, long-​
possible bronchiectasis exacerbations187 term gentamicin therapy provides substantial benefit in
• Reducing overcrowding: overcrowding is a risk factor for ARIs203 bronchiectasis, but ongoing efficacy requires continu-
• Vaccinations (influenza and pneumococcal): prevent early and/or recurrent ARIs. ous treatment, as clinical parameters deteriorated after
Reduction in the prevalence of bronchiectasis with childhood vaccines (for example, stopping the drug, during the 3 months off-​treatment
measles and pertussis)203 follow-​up period167. This relapse is probably explained
• Optimizing nutrition: poor nutrition is associated with defective immune function by the fact that studies consistently show that inhaled
and susceptibility to infections203. Vitamin D supplementation protects against ARIs, antibiotics do not completely eradicate the infection and
especially those who have deficiency107 bacterial loads return to baseline levels within 1 month
• Early treatment of chronic wet or productive cough: reduces the effects of chronic of discontinuing the drugs.
airway infection and inflammation; chronic cough duration correlates with reduced Efficacy of aztreonam solution for inhalation was
lung function and worse radiology bronchiectasis scores in children and adults. Early assessed in two double-​blind, multicentre, placebo-​
treatment arrests processes leading to bronchiectasis47,204 controlled phase III trials designated AIR-​BX1 and
• Early detection of aetiologies that can lead to bronchiectasis: in children at risk of AIR-​BX2 (ref.159). The primary outcome of both studies
bronchiectasis, early treatment of their risk factor (such as ARIs) prevents the was the QOL score, as measured by a newly developed
development of pneumonia and bronchiectasis bronchiectasis-​specific QOL measure: the QOL ques-
• Improving oral health: reduces aspiration of oral bacteria and, therefore, reduces tionnaire bronchiectasis respiratory symptoms score
ARIs. Improving oral hygiene reduced pneumonia-​related mortality and non-​fatal (QOL-​B-RSS)159. The difference between aztreonam
episodes in hospitalized elderly people and nursing home residents205 and placebo for the adjusted mean QOL-​B-RSS change
• Improving socioeconomic determinants: bronchiectasis has a higher prevalence in from baseline score was not clinically meaningful in
less-​affluent settings206. Low socioeconomic status is associated with chronic either AIR-​BX1 or AIR-​BX2 studies. Increased adverse
respiratory disease
effects were observed in the aztreonam arm compared

12 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

with placebo, including dyspnoea, cough and increased reduce neutrophilic inflammation by reducing neutro-
sputum. In conclusion, aztreonam did not provide sub- phil recruitment to the lung, for example, with antago­
stantial benefit in patients with bronchiectasis in this nists of C-​X-C chemo­kine receptor type 2 (CXCR2).
trials; by contrast, aztreonam therapy is successful in This approach did reduce sputum neutrophil count
cystic fibrosis, suggesting the need for further placebo-​ but seemed to increase the risk of infections in a small
controlled studies to establish the clinical benefit of this randomized controlled trial171. Approaches that aim to
inhaled antibiotic159. reduce neutrophil numbers are unlikely to be success-
Two inhaled ciprofloxacin preparations have recently ful, as neutrophils are required for the control of air-
been evaluated: a dry powder formulation and a lipo- way infection — removing these cells leads to increased
somal formulation. Two randomized controlled trials infections, as demonstrated by a trial of a leukotriene
(RESPIRE 1 and RESPIRE 2) of dry powder ciprofloxa- B4 antagonist in patients with cystic fibrosis, in which
cin failed to show consistent significant benefits in terms the drug increased the number of infections and caused
of the primary outcomes of frequency of exacerbations FEV1 decline175. Alternative approaches that modulate
or time to first exacerbation160–162. Two trials of cyclical rather than inhibit inflammatory functions may be
treatment with ARD-3150 (an inhaled formulation of more successful. Inhibitors of neutrophil elastase have
ciprofloxacin containing liposome-​encapsulated cipro- been tested in a small proof-​of-concept study and a
floxacin and free ciprofloxacin) also had inconsistent significant improvement in FEV1 was observed, with
results in terms of their primary end point of time to first trends towards improvement in QOL, but larger studies
exacerbation, with one trial showing a significant pro- are required172.
longation of time to first exacerbation but no significant
difference in the replicate trial. Pooled data from both Surgery and transplantation
trials showed a significant reduction in the more-​relevant Although surgical resection of affected lung sections was
clinical end point of frequency of exacerbation 168. used extensively in the past, its role is now limited to
Treatment was safe and well tolerated. severe localized bronchiectasis in which medical man-
In conclusion, inhaled antibiotics may be beneficial agement has failed or to specific complications, such
in some patients with bronchiectasis, but the optimal as severe haemoptysis36. In a meta-​analysis of carefully
patient population has not been identified. Although selected patients with localized bronchiectasis and who
the ability of antibiotics to reduce bacterial load has were assessed as being suitable for surgery, the pooled
been demonstrated for all inhaled antibiotics, this effect mortality was 1.4%, with a postoperative morbidity
is not consistently associated with an improvement in rate of 16.2%. Surgery completely alleviated symptoms
symptoms or exacerbations. in 71.5% of patients176. Close collaboration between
expert physicians and surgeons is mandatory if surgery
Inhaled corticosteroids and inhaled bronchodilators is considered.
Whereas inhaled corticosteroids and bronchodilators The success of medical therapy means that bronchiec-
have an established role in the management of asthma tasis is now an infrequent indication for lung transplan-
and COPD169, their use in bronchiectasis is currently tation. A report from a UK transplant centre identified
controversial. The ERS and the Thoracic Society of 42 patients with bronchiectasis receiving transplanta-
Australia and New Zealand guidelines suggest consider- tion over a 23-year period. Consistent with indication
ing bronchodilators in patients with bronchiectasis with for transplant in other respiratory diseases, the major
symptoms of breathlessness or features consistent characteristics of this bronchiectasis cohort were severe
with asthma36,108. Both guidelines recommend against lung function impairment (mean 22% FEV1 predicted)
the use of inhaled corticosteroids, except in patients with severe disability. Outcomes were favourable, with
with coexisting asthma or COPD; nevertheless, inhaled survival of 74% at 1 year and 48% at 10 years177.
corticosteroids are commonly used in clinical practice.
An updated Cochrane review of seven randomized con- Quality of life
trolled trials in adults with bronchiectasis found that Daily symptoms and fatigue, shortness of breath and
inhaled corticosteroids did not improve lung function, productive cough with unexpected exacerbations are a
exacerbation frequency or QOL170. detriment to the physical and mental health of patients
with bronchiectasis178,179. Symptoms also interfere with
Anti-​inflammatories both work and social events; in particular, the produc-
Although bronchiectasis is an inflammatory disorder, tive cough is often reported by patients to be publicly
our understanding of the inflammatory processes lead- embarrassing. Finally, the progressive and incurable
ing to bronchiectasis is limited, and this is probably nature of bronchiectasis is a source of stress and anx­
the reason why anti-​inflammatory therapy has been iety180. Medical care utilization and expenditures are also
unsuccessful to date171–174. In a randomized controlled a burden for patients with bronchiectasis, as they incur
trial assessing the role of atorvastatin in adult patients more frequent outpatient visits, longer hospitalizations
with clinically significant bronchiectasis and ≥2 exacer­ and more antibiotic therapy than age-​matched controls
bations in the past year, high-​dose atorvastatin was with other chronic diseases (for example, diabetes mel-
associated with a significant improvement in cough174. litus, coronary disease and HIV infection)181. Of note,
However, in a similar trial in patients with P. aeruginosa bronchiectasis also imposes a substantial burden on
infection, there was no improvement in cough173. Many caretakers of patients; compared with the general popu­
anti-​inflammatory approaches have attempted to lation, mothers of children with bronchiectasis report

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 13

0123456789();
P­r i­m e­r

increased anxiety, stress and depression, which worsen modern ‘omics’ technologies is beginning to identify
during exacerbations and are independent of the severity key pathological and microbial changes. Although
of disease182. 20–40% of patients are still diagnosed with idiopathic
Patient-​reported outcome (PRO) questionnaires are bronchiectasis15, the increasing availability of exome and
self-​administered tools that measure the HRQOL from whole-​genome sequencing is enabling the identification
the perspective of the patient. PRO questionnaires can be of genetic variants that cause bronchiectasis, including
completed at different time points to measure the effect PIK3CD mutations that cause the primary immuno-
of an intervention (for example, an antibiotic), event (for deficiency known as activated PI3Kδ syndrome 186.
example, exacerbation) or disease feature (for example, A key question is how the immune response becomes
the presence of P. aeruginosa infection). In contrast to dysregulated in bronchiectasis. Airway innate immune
diseases (such as hypertension or diabetes mellitus) that responses are rapid and respond to foreign invasion
can be monitored by parameters that can be measured in tandem with pathogen-​specific adaptive responses. In
objectively, bronchiectasis symptoms are overall sub- healthy individuals, inflammation resolves and the patho­
jective. Thus, HRQOL measurements through PRO gen is cleared; however, in bronchiectasis, an exagger-
questionnaires have enabled clinicians and research- ated response, coupled to resolution and repair failure,
ers to measure the adverse effects of these subjective results in the accrual of a harmful mixture of mucus,
symptoms and gauge which ones are most important to pathogens and immune cells in the airway, further
the patient. The US FDA and the European Medicines damaging host tissue and creating an environment that
Agency embrace the use of PRO measures. Several PRO favours perpetuation of the pathogenetic mechanisms.
questionnaires have been validated and used as primary
and secondary outcomes in clinical trials of bronchiecta- Patient selection
sis drugs. The QOL-​B questionnaire and the St George’s One of the greatest challenges in bronchiectasis, as out-
Respiratory Questionnaire can be used to assess overall lined in this Primer, is that the majority of clinical trials
QOL, and the Parent-​proxy cough-​specific QOL and the of this condition have failed. Mortality is not a useful
Leicester cough questionnaire can be used to assess clinical end point for bronchiectasis trials, and more-​
the effect of cough183. relevant clinical end points that could be modified with
Small studies investigating the effect of exacerbations therapy (such as the frequency of pulmonary exacerba-
on patients with bronchiectasis demonstrate a measur­able tions and hospital admissions and QOL) could better
decrement in HRQOL scores during exacerbations184. reflect the severity of disease43,160–162,187. The failure of
After treatment with an antibiotic, HRQOL improves previous clinical trials in patients with bronchiectasis
beyond steady-​state scores159,184 and remains above could also reflect an inability to correctly select those
steady-​state levels even when inflammatory markers patients who are most likely to benefit from the treat-
rise again185. However, in large clinical trials of inhaled ment (for example, those most likely to experience an
antibiotics, HRQOL performance has been inconsist- exacerbation or whose underlying aetiology is likely
ent. For example, in the RESPIRE trials160–162, the treat- to respond to that specific treatment)43. To address
ment arms that showed reduction in exacerbations did this issue, large international registries, such as the
not measure improvement in HRQOL scores. In some European Bronchiectasis Registry at the EMBARC, are
instances, the St George’s Respiratory Questionnaire and providing key data on the natural history of bronchiec-
QOL-B questionnaire yielded discordant results160,161. tasis that will be required to validate clinical trial end
The under­lying reason for these varying results probably points, such as QOL and exacerbations10,41,178,188. The
represents more than a difference in medications. Rather, identification of clinical phenotypes (groups of patient
it may be the heterogeneous nature of the population of characteristics that are associated with a specific clini­
patients with bronchiectasis1. In addition, the influence cal outcome) will help to selectively include in trials
of socioeconomic and/or ethnic differences on HRQOL the patients who are more likely to have bronchiectasis-​
has not yet been addressed in bronchiectasis. There are associated events for example, the frequent exacerbator
currently no FDA-​approved therapies for bronchiec- phenotype includes patients who consistently exacer-
tasis, in part because HRQOL results are mixed, and bate over time and have poor clinical outcomes as a
the lack of approved therapies probably compounds the result34,189,190. However, multiple underlying biological
HRQOL problem. processes may be responsible for a specific pheno-
type34,74,191,192. Endotyping refers to identifying under­
Outlook lying biological processes that can be measured and are
Several questions in bronchiectasis, from pathophysio­ associated with specific clinical outcomes or treatment
logy to long-​term management, remain unanswered; in response192,193. Examples of candidate endotypes include
2016, the EMBARC published a list of 55 key research patients with increased neutrophil elastase levels in spu-
priorities identified from a survey of bronchiectasis tum, who have worse clinical outcomes with faster lung
experts and >700 patients from across Europe8. function decline and may theor­etically be more likely
to respond to antibiotic treatment or to direct inhibi-
Pathophysiology tion of elastase77. Other endotypes are represented by
Research into the pathophysiology of the disease is patients with elevated airway bacterial load, who may
greatly limited by the lack of an available animal model be more likely to respond to inhaled antibiotics, or those
that reproduces key features of the syndrome; however, with increased blood eosinophil counts, who may be
the study of patients with early bronchiectasis using more likely to respond to inhaled cortico­steroids194–196.

14 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

The concepts of phenotypes and endotypes require is required and will be achieved by the development of
prospective testing in trials. biomarkers and stratification tools that can be used in
clinical practice and preferably at the point of care43,192,193.
Management Finally, therapeutic development in bronchiectasis
Multiple new therapies are in development for bronchi- has, until now, focused exclusively on end-​stage disease.
ectasis, including new antibiotics, but also new appro­ However, bronchiectasis and even ‘pre-​bronchiectasis’
aches, such as inhibition of dipeptidyl peptidase 1, (in which the clinical syndrome is evident before the
which activates neutrophil serine proteases in the bone development of radiological bronchial dilatation) can
marrow197. Drugs designed to improve mucociliary be recognized early, and the airway dilatation that
clearance include CFTR modulators or correctors and defines bronchiectasis is likely to be a consequence of
epithelial sodium channel inhibitors43, whereas immuno- the under­lying pathology rather than the cause of the
modulators currently being tested include granulocyte– disease. Patients with an initial inflammatory process
macrophage colony-​stimulating factor, neutrophil associated with airway infection and neutrophilia can
elastase inhibitors, roflumilast (a phosphodiesterase increasingly be recognized and treated before airway
type 4 inhibitor) and vitamin D supplementation66,107. dilatation develops. Prevention is better than cure, and
The possible role of these therapies has recently been ultimately, the solution to the increase in bronchiectasis
extensively reviewed43. prevalence may be to refocus efforts towards effective
New pathogenetic pathways, particularly of inflam- recognition and prevention.
matory and ciliary dysfunction, are being identified and In summary, bronchiectasis is a common, disabling
are likely to lead to many new therapeutic opportunities. chronic lung disease caused by a wide range of under-
Unravelling the heterogeneity of bronchiectasis is the lying disorders. The renewed interest in bronchiectasis
key research priority in bronchiectasis, as none of the has brought new understanding to its pathophysiology,
therapies in current development is likely to be appropri- new clinical trials and better ways of evaluating QOL
ate for all patients, and even antibiotics, which have been and patient prognosis. All clinicians should be aware of
the mainstay of therapy since their widespread availa- the principles of managing this disorder in adults and
bility in the 1950s, are only required for some patients children, and scientists should view this disease as a field
and their use in patients who do not need them could ripe for new discoveries in pathogenesis and therapy.
contribute to the development of antibiotic resistance
without giving clinical benefits. Personalized medicine Published online xx xx xxxx

1. Chalmers, J. D., Aliberti, S. & Blasi, F. Management 11. Aksamit, T. R. et al. Adult patients with bronchiectasis: 23. Singleton, R. et al. Bronchiectasis in Alaska native
of bronchiectasis in adults. Eur. Respir. J. 45, a first look at the US Bronchiectasis Research Registry. children: causes and clinical courses. Pediatr.
1446–1462 (2015). Chest 151, 982–992 (2017). Pulmonol. 29, 182–187 (2000).
2. Cole, P. J. Inflammation: a two-​edged sword — the The first report from a national registry in the 24. Chandrasekaran, R., Mac Aogain, M., Chalmers, J. D.,
model of bronchiectasis. Eur. J. Respir. Dis. Suppl. United States demonstrating patient characteristics Elborn, S. J. & Chotirmall, S. H. Geographic variation
147, 6–15 (1986). very different from European and Australasian in the aetiology, epidemiology and microbiology of
3. Chalmers, J. D. et al. The bronchiectasis severity cohorts, including very high rates of NTM infection. bronchiectasis. BMC Pulm. Med. 18, 83 (2018).
index. An international derivation and validation 12. Taylor-​Cousar, J. L. et al. Tezacaftor-​ivacaftor in patients 25. Weycker, D., Hansen, G. L. & Seifer, F. D. Prevalence
study. Am. J. Respir. Crit. Care Med. 189, 576–585 with cystic fibrosis homozygous for Phe508del. N. Engl. and incidence of noncystic fibrosis bronchiectasis
(2014). J. Med. 377, 2013–2023 (2017). among US adults in 2013. Chron. Respir. Dis. 14,
This study described the risk factors for mortality 13. Hess, E. P. et al. Trends in computed tomography 377–384 (2017).
and hospital admissions in multiple European utilization rates: a longitudinal practice-​based study. 26. Lin, J.-L., Xu, J.-F. & Qu, J.-M. Bronchiectasis in China.
bronchiectasis cohorts and derived a clinical J. Patient Saf. 10, 52–58 (2014). Ann. Am. Thorac Soc. 13, 609–616 (2016).
prediction tool that can predict poor outcome 14. Blackall, S. R. et al. Bronchiectasis in indigenous 27. McDonnell, M. J. et al. Comorbidities and the risk of
across multiple domains, which has now been and non-​indigenous residents of Australia and mortality in patients with bronchiectasis: an
validated worldwide. New Zealand. Respirology 23, 743–749 (2018). international multicentre cohort study. Lancet Respir.
4. Tomos, I., Karakatsani, A., Manali, E. D. & Papiris, S. A. 15. Lonni, S. et al. Etiology of non-​cystic fibrosis Med. 4, 969–979 (2016).
Celebrating two centuries since the invention of the bronchiectasis in adults and its correlation to disease 28. Diel, R. et al. Burden of non-​tuberculous
stethoscope. Rene Theophile Hyacinthe Laënnec severity. Ann. Am. Thorac Soc. 12, 1764–1770 mycobacterial pulmonary disease in Germany.
(1781–1826). Ann. Am. Thorac Soc. 13, 1667–1670 (2015). Eur. Respir. J. 49, 1602109 (2017).
(2016). 16. Seitz, A. E., Olivier, K. N., Adjemian, J., Holland, S. M. 29. Koh, W.-J. et al. Outcomes of Mycobacterium avium
5. Goeminne, P. C. & De Soyza, A. Bronchiectasis: how to & Prevots, D. R. Trends in bronchiectasis among complex lung disease based on clinical phenotype.
be an orphan with many parents? Eur. Respir. J. 47, Medicare beneficiaries in the United States, 2000 to Eur. Respir. J. 50, 1602503 (2017).
10–13 (2016). 2007. Chest 142, 432–439 (2012). 30. Vidaillac, C., Yong, V. F. L., Jaggi, T. K., Soh, M.-M. &
6. Quint, J. K. et al. Changes in the incidence, prevalence 17. Ringshausen, F. C. et al. Bronchiectasis-​associated Chotirmall, S. H. Gender differences in bronchiectasis:
and mortality of bronchiectasis in the UK from 2004 hospitalizations in Germany, 2005–2011: a a real issue? Breathe (Sheff.) 14, 108–121 (2018).
to 2013: a population-​based cohort study. Eur. Respir. J. population-​based study of disease burden and trends. 31. Vogelmeier, C. F. et al. Global strategy for the
47, 186–193 (2016). PLOS ONE 8, e71109 (2013). diagnosis, management, and prevention of chronic
An epidemiological study using routinely collected 18. Sanchez-​Munoz, G. et al. Time trends in hospital obstructive lung disease 2017 report: GOLD executive
data, demonstrating dramatic increases in admissions for bronchiectasis: analysis of the Spanish summary. Am. J. Respir. Crit. Care Med. 195,
bronchiectasis prevalence over 10 years. National Hospital discharge data. PLOS ONE 11, 557–582 (2017).
7. Ringshausen, F. C. et al. Bronchiectasis in Germany: e0162282 (2016). 32. Tan, W. C. et al. Findings on thoracic computed
a population-​based estimation of disease prevalence. 19. Eastham, K. M., Fall, A. J., Mitchell, L. & Spencer, D. A. tomography scans and respiratory outcomes in
Eur. Respir. J. 46, 1805–1807 (2015). The need to redefine non-​cystic fibrosis bronchiectasis in persons with and without chronic obstructive
8. Aliberti, S. et al. Research priorities in bronchiectasis: childhood. Thorax 59, 324–327 (2004). pulmonary disease: a population-​based cohort study.
a consensus statement from the EMBARC Clinical 20. Karakoc, G. B., Yilmaz, M., Altintas, D. U. & PLOS ONE 11, e0166745 (2016).
Research Collaboration. Eur. Respir. J. 48, 632–647 Kendirli, S. G. Bronchiectasis: still a problem. 33. De Soyza, A. et al. Bronchiectasis rheumatoid overlap
(2016). Pediatr. Pulmonol. 32, 175–178 (2001). syndrome is an independent risk factor for mortality in
9. McDonnell, M. J. et al. Multidimensional severity 21. Twiss, J., Metcalfe, R., Edwards, E. & Byrnes, C. patients with bronchiectasis: a multicenter cohort
assessment in bronchiectasis: an analysis of New Zealand national incidence of bronchiectasis study. Chest 151, 1247–1254 (2017).
seven European cohorts. Thorax 71, 1110–1118 “too high” for a developed country. Arch. Dis. Child 90, 34. Chalmers, J. D. et al. Characterization of the “frequent
(2016). 737–740 (2005). exacerbator phenotype” in bronchiectasis. Am. J.
10. Chalmers, J. D. et al. The EMBARC European 22. Chang, A. B., Grimwood, K., Mulholland, E. K. & Respir. Crit. Care Med. 197, 1410–1420 (2018).
Bronchiectasis Registry: protocol for an international Torzillo, P. J. Bronchiectasis in Indigenous children in A large (>2,000 patients), multicentre study
observational study. ERJ Open Res. https://doi.org/ remote Australian communities. Med. J. Aust. 177, describing the clinical effect of exacerbations over
10.1183/23120541.00081-2015 (2016). 200–204 (2002). time, demonstrating that patients with frequent

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 15

0123456789();
P­r i­m e­r

exacerbations have worse clinical outcomes even 55. Flume, P. A., Chalmers, J. D. & Olivier, K. N. Advances idiopathic pulmonary fibrosis. Respiration 67, 52–59
after adjustment for potential confounders. in bronchiectasis: endotyping, genetics, microbiome, (2000).
35. Araujo, D. et al. The independent contribution of and disease heterogeneity. Lancet 392, 880–890 80. Fick, R. B. J. et al. Proteins of the cystic fibrosis
Pseudomonas aeruginosa infection to long-​term (2018). respiratory tract. Fragmented immunoglobulin G
clinical outcomes in bronchiectasis. Eur. Respir. J. 51, 56. Taylor, S. L. et al. Matrix metalloproteinases vary with opsonic antibody causing defective opsonophagocytosis.
1701953 (2018). airway microbiota composition and lung function in J. Clin. Invest. 74, 236–248 (1984).
36. Polverino, E. et al. European Respiratory Society non-​cystic fibrosis bronchiectasis. Ann. Am. Thorac Soc. 81. Tosi, M. F., Zakem, H. & Berger, M. Neutrophil
guidelines for the management of adult bronchiectasis. 12, 701–707 (2015). elastase cleaves C3bi on opsonized pseudomonas as
Eur. Respir. J. 50, 1700629 (2017). 57. Chintalacharuvu, K. R. et al. Cleavage of the human well as CR1 on neutrophils to create a functionally
The first international guidelines following on immunoglobulin A1 (IgA1) hinge region by IgA1 important opsonin receptor mismatch. J. Clin. Invest.
from national guidance issued by thoracic societies proteases requires structures in the Fc region of IgA. 86, 300–308 (1990).
in Spain, the United Kingdom, Australia and Infect. Immun. 71, 2563–2570 (2003). 82. Berger, M., Sorensen, R. U., Tosi, M. F., Dearborn, D. G.
New Zealand. 58. Wells, T. J. et al. Increased severity of respiratory & Doring, G. Complement receptor expression on
37. Mao, B., Yang, J.-W., Lu, H.-W. & Xu, J.-F. Asthma and infections associated with elevated anti-​LPS IgG2 neutrophils at an inflammatory site, the
bronchiectasis exacerbation. Eur. Respir. J. 47, which inhibits serum bactericidal killing. J. Exp. Med. Pseudomonas-​infected lung in cystic fibrosis.
1680–1686 (2016). 211, 1893–1904 (2014). J. Clin. Invest. 84, 1302–1313 (1989).
38. Shah, P. L. et al. Determinants of chronic infection with 59. van Ingen, J. et al. Treatment outcome definitions in 83. Voglis, S. et al. Human neutrophil peptides and
Staphylococcus aureus in patients with bronchiectasis. nontuberculous mycobacterial pulmonary disease: phagocytic deficiency in bronchiectatic lungs.
Eur. Respir. J. 14, 1340–1344 (1999). an NTM-​NET consensus statement. Eur. Respir. J. 51, Am. J. Respir. Crit. Care Med. 180, 159–166 (2009).
39. Whitehouse, J. L., Exley, A. R., Foweraker, J. & Bilton, D. 1800170 (2018). The most detailed study of neutrophil function
Chronic Burkholderia multivorans bronchial infection in 60. Kunst, H., Wickremasinghe, M., Wells, A. & Wilson, R. and dysfunction in bronchiectasis to date,
a non-​cystic fibrosis individual with mannose binding Nontuberculous mycobacterial disease and demonstrating that human neutrophil peptides
lectin deficiency. Thorax 60, 168–170 (2005). Aspergillus-​related lung disease in bronchiectasis. (defensins) impair neutrophil phagocytosis and
40. Shoemark, A. et al. High prevalence of CCDC103 p. Eur. Respir. J. 28, 352–357 (2006). contribute to airway inflammation in bronchiectasis.
His154Pro mutation causing primary ciliary dyskinesia 61. Mac Aogain, M. et al. Immunological corollary of the 84. Weldon, S. et al. Decreased levels of secretory
disrupts protein oligomerisation and is associated pulmonary mycobiome in bronchiectasis: the CAMEB leucoprotease inhibitor in the Pseudomonas-​infected
with normal diagnostic investigations. Thorax 73, study. Eur. Respir. J. 52, 1800766 (2018). cystic fibrosis lung are due to neutrophil elastase
157–166 (2018). 62. Gao, Y.-H. et al. The role of viral infection in pulmonary degradation. J. Immunol. 183, 8148–8156 (2009).
41. Chalmers, J. D. et al. The European Multicentre exacerbations of bronchiectasis in adults: a 85. Amitani, R. et al. Effects of human neutrophil elastase
Bronchiectasis Audit and Research Collaboration prospective study. Chest 147, 1635–1643 (2015). and Pseudomonas aeruginosa proteinases on human
(EMBARC): experiences from a successful ERS 63. Chalmers, J. D. & Hill, A. T. Mechanisms of immune respiratory epithelium. Am. J. Respir. Cell Mol. Biol. 4,
Clinical Research Collaboration. Breathe (Sheff.) 13, dysfunction and bacterial persistence in non-​cystic 26–32 (1991).
180–192 (2017). fibrosis bronchiectasis. Mol. Immunol. 55, 27–34 86. Fischer, B. M. & Voynow, J. A. Neutrophil elastase
42. Donnelly, D., Critchlow, A. & Everard, M. L. Outcomes (2013). induces MUC5AC gene expression in airway epithelium
in children treated for persistent bacterial bronchitis. 64. Saleh, A. D., Kwok, B., Brown, J. S. & Hurst, J. R. via a pathway involving reactive oxygen species.
Thorax 62, 80–84 (2007). Correlates and assessment of excess cardiovascular Am. J. Respir. Cell Mol. Biol. 26, 447–452 (2002).
43. Chalmers, J. D. & Chotirmall, S. H. Bronchiectasis: risk in bronchiectasis. Eur. Respir. J. 50, 1701127 87. Bedi, P. et al. The BRICS (Bronchiectasis Radiologically
new therapies and new perspectives. Lancet Respir. (2017). Indexed CT Score): a multicenter study score for use in
Med. 6, 715–726 (2018). 65. Frija-​Masson, J. et al. Bacteria-​driven peribronchial idiopathic and postinfective bronchiectasis. Chest
44. Chalmers, J. D. et al. Short- and long-​term antibiotic lymphoid neogenesis in bronchiectasis and cystic 153, 1177–1186 (2018).
treatment reduces airway and systemic inflammation fibrosis. Eur. Respir. J. 49, 1601873 (2017). 88. Gaga, M. et al. Increases in CD4+ T lymphocytes,
in non-​cystic fibrosis bronchiectasis. Am. J. Respir. 66. Ruchaud-​Sparagano, M.-H. et al. Effect of macrophages, neutrophils and interleukin 8 positive
Crit. Care Med. 186, 657–665 (2012). granulocyte–macrophage colony-​stimulating factor cells in the airways of patients with bronchiectasis.
A study demonstrating that bacterial clearance on neutrophil function in idiopathic bronchiectasis. Thorax 53, 685–691 (1998).
through antibiotic therapy results in reduced airway Respirology 18, 1230–1235 (2013). 89. Zheng, L. et al. Macrophages, neutrophils and tumour
inflammation and in improvements in systemic 67. van de Ven, A. A. J. M. et al. A CT scan score for the necrosis factor-​alpha expression in bronchiectatic
inflammation, thereby providing strong support for assessment of lung disease in children with common airways in vivo. Respir. Med. 95, 792–798 (2001).
the self-​perpetuating cycle hypothesis and the variable immunodeficiency disorders. Chest 138, 90. Hodge, S. et al. Is alveolar macrophage phagocytic
development of inhaled antibiotics as a treatment. 371–379 (2010). dysfunction in children with protracted bacterial
45. Finch, S., McDonnell, M. J., Abo-​Leyah, H., Aliberti, S. 68. Frick, A. G. et al. Haemophilus influenzae stimulates bronchitis a forerunner to bronchiectasis? Chest 149,
& Chalmers, J. D. A. Comprehensive analysis of the ICAM-1 expression on respiratory epithelial cells. 508–515 (2016).
impact of Pseudomonas aeruginosa colonization on J. Immunol. 164, 4185–4196 (2000). A mechanistic study that increases our
prognosis in adult bronchiectasis. Ann. Am. Thorac 69. Angrill, J. et al. Bronchial inflammation and colonization understanding of persistent bacterial bronchitis in
Soc. 12, 1602–1611 (2015). in patients with clinically stable bronchiectasis. Am. J. children by demonstrating reductions in alveolar
46. Wurzel, D. F. et al. Protracted bacterial bronchitis in Respir. Crit. Care Med. 164, 1628–1632 (2001). macrophage phagocytosis and efferocytosis
children: natural history and risk factors for 70. Saleh, A. D. et al. The heterogeneity of systemic (the clearance of apoptotic cells) in children with
bronchiectasis. Chest 150, 1101–1108 (2016). inflammation in bronchiectasis. Respir. Med. 127, persistent bacterial bronchitis compared with
A detailed description of a cohort of patients with 33–39 (2017). healthy controls, suggesting a defect that may
persistent bacterial bronchitis that also identified 71. van Kessel, D. A., van Velzen-​Blad, H., van den Bosch, contribute to persistent neutrophilic inflammation.
risk factors for progression to bronchiectasis. J. M. M. & Rijkers, G. T. Impaired pneumococcal 91. Vandivier, R. W. et al. Elastase-​mediated
47. Kantar, A. et al. ERS statement on protracted bacterial antibody response in bronchiectasis of unknown phosphatidylserine receptor cleavage impairs
bronchitis in children. Eur. Respir. J. 50, 1602139 aetiology. Eur. Respir. J. 25, 482–489 (2005). apoptotic cell clearance in cystic fibrosis and
(2017). 72. Zimmer, J., Sleiman, M., Poli, A., Michel, T. & bronchiectasis. J. Clin. Invest. 109, 661–670 (2002).
A consensus statement from the ERS on the Hentges, F. TAP deficiency is also a cause of 92. Houtmeyers, E., Gosselink, R., Gayan-​Ramirez, G. &
definition, diagnosis, microbiology, treatment bronchiectasis. Thorax 68, 490–491 (2013). Decramer, M. Regulation of mucociliary clearance in
and research priorities in persistent bacterial 73. Holmes, A. H., Pelton, S., Steinbach, S. & Luzzi, G. A. health and disease. Eur. Respir. J. 13, 1177–1188
bronchitis. HIV related bronchiectasis. Thorax 50, 1227 (1995). (1999).
48. Faner, R. et al. The microbiome in respiratory 74. Tsikrika, S. et al. The role of non-​invasive modalities for 93. Gilley, S. K. et al. Deletion of airway cilia results in
medicine: current challenges and future perspectives. assessing inflammation in patients with non-​cystic noninflammatory bronchiectasis and hyperreactive
Eur. Respir. J. 49, 1602086 (2017). fibrosis bronchiectasis. Cytokine 99, 281–286 (2017). airways. Am. J. Physiol. Lung Cell. Mol. Physiol. 306,
49. Taylor, S. L. et al. FUT2 genotype influences lung 75. Watt, A. P. et al. Neutrophil apoptosis, L162–L169 (2014).
function, exacerbation frequency and airway microbiota proinflammatory mediators and cell counts in 94. Smallman, L. A., Hill, S. L. & Stockley, R. A. Reduction
in non-​CF bronchiectasis. Thorax 72, 304–310 (2017). bronchiectasis. Thorax 59, 231–236 (2004). of ciliary beat frequency in vitro by sputum from
50. Metersky, M. L. et al. The prevalence and significance 76. Chalmers, J. D. et al. Neutrophil elastase activity is patients with bronchiectasis: a serine proteinase
of Staphylococcus aureus in patients with non-​cystic associated with exacerbations and lung function effect. Thorax 39, 663–667 (1984).
fibrosis bronchiectasis. Ann. Am. Thorac Soc. 15, decline in bronchiectasis. Am. J. Respir. Crit. Care Med. 95. Shoemark, A. et al. Primary ciliary dyskinesia with
365–370 (2018). 195, 1384–1393 (2017). normal ultrastructure: three-​dimensional tomography
51. Hilliam, Y. et al. Pseudomonas aeruginosa adaptation A study demonstrating for the first time that a detects absence of DNAH11. Eur. Respir. J. 51,
and diversification in the non-​cystic fibrosis biomarker could predict clinical outcomes in 1701809 (2018).
bronchiectasis lung. Eur. Respir. J. 49, 1602108 bronchiectasis, supporting a key role of neutrophil 96. Falconer, M., Collins, D. R., Feeney, J. &
(2017). elastase in disease progression. Torreggiani, W. C. Mounier–Kuhn syndrome in an
52. Woo, T. E. et al. Virulence adaptations of 77. Schleimer, R. P., Benenati, S. V., Friedman, B. & older patient. Age Ageing 37, 115–116 (2008).
Pseudomonas aeruginosa isolated from patients with Bochner, B. S. Do cytokines play a role in leukocyte 97. Nishino, M. et al. Excessive collapsibility of bronchi in
non-​cystic fibrosis bronchiectasis. Microbiology 162, recruitment and activation in the lungs? Am. Rev. bronchiectasis: evaluation on volumetric expiratory
2126–2135 (2016). Respir. Dis. 143, 1166–1169 (1991). high-​resolution CT. J. Comput. Assist. Tomogr. 30,
53. King, P. T. et al. Adaptive immunity to nontypeable 78. Mikami, M., Llewellyn-​Jones, C. G., Bayley, D., Hill, S. L. 474–478 (2006).
Haemophilus influenzae. Am. J. Respir. Crit. Care Med. & Stockley, R. A. The chemotactic activity of sputum 98. Lucas, J. S. et al. European Respiratory Society
167, 587–592 (2003). from patients with bronchiectasis. Am. J. Respir. Crit. guidelines for the diagnosis of primary ciliary
54. Walker, W. T. et al. Primary ciliary dyskinesia ciliated Care Med. 157, 723–728 (1998). dyskinesia. Eur. Respir. J. 49, 1601090 (2017).
airway cells show increased susceptibility to 79. Schaaf, B., Wieghorst, A., Aries, S. P., Dalhoff, K. & 99. Goutaki, M. et al. The international primary ciliary
Haemophilus influenzae biofilm formation. Eur. Respir. J. Braun, J. Neutrophil inflammation and activation in dyskinesia cohort (iPCD cohort): methods and first
50, 1700612 (2017). bronchiectasis: comparison with pneumonia and results. Eur. Respir. J. 49, 1601181 (2017).

16 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();
P­r i­m e­r

100. Driscoll, J. A., Bhalla, S., Liapis, H., Ibricevic, A. & multidetector CT acquisition in the assessment of aerosolized recombinant human DNase I. rhDNase
Brody, S. L. Autosomal dominant polycystic kidney bronchiectasis. Br. J. Radiol. 83, 67–70 (2010). Study Group. Chest 113, 1329–1334 (1998).
disease is associated with an increased prevalence of 125. Dodd, J. D., Souza, C. A. & Muller, N. L. Conventional One of the first large-​scale trials in bronchiectasis
radiographic bronchiectasis. Chest 133, 1181–1188 high-​resolution CT versus helical high-​resolution MDCT in the modern era, showing that a drug known to
(2008). in the detection of bronchiectasis. AJR Am. J. be beneficial in cystic fibrosis bronchiectasis was
101. Fajac, I., Viel, M., Gaitch, N., Hubert, D. & Bienvenu, T. Roentgenol. 187, 414–420 (2006). ineffective or potentially harmful in non-​cystic
Combination of ENaC and CFTR mutations may 126. Goyal, V., Grimwood, K., Marchant, J. M., Masters, I. B. fibrosis bronchiectasis; the knowledge that
predispose to cystic fibrosis-​like disease. Eur. Respir. J. & Chang, A. B. Paediatric chronic suppurative lung cystic fibrosis and bronchiectasis behave differently
34, 772–773 (2009). disease: clinical characteristics and outcomes. Eur. J. has made a major impact on the field.
102. Casals, T. et al. Bronchiectasis in adult patients: Pediatr. 175, 1077–1084 (2016). 150. Wills, P. J. et al. Short-​term recombinant human
an expression of heterozygosity for CFTR gene 127. Colom, A. J., Maffey, A., Garcia Bournissen, F. & DNase in bronchiectasis. Effect on clinical state and
mutations? Clin. Genet. 65, 490–495 (2004). Teper, A. Pulmonary function of a paediatric cohort in vitro sputum transportability. Am. J. Respir. Crit.
103. Bienvenu, T. et al. Cystic fibrosis transmembrane of patients with postinfectious bronchiolitis Care Med. 154, 413–417 (1996).
conductance regulator channel dysfunction in non-​cystic obliterans. A long term follow-​up. Thorax 70, 151. Mustafa, M.-H. et al. Acquired resistance to
fibrosis bronchiectasis. Am. J. Respir. Crit. Care Med. 169–174 (2015). macrolides in Pseudomonas aeruginosa from
181, 1078–1084 (2010). 128. Pizzutto, S. J. et al. Bronchoscopy contributes to the cystic fibrosis patients. Eur. Respir. J. 49, 1601847
104. Horvath, I. et al. Increased levels of exhaled carbon clinical management of Indigenous children newly (2017).
monoxide in bronchiectasis: a new marker of oxidative diagnosed with bronchiectasis. Pediatr. Pulmonol. 48, 152. Kelly, C. et al. Macrolide antibiotics for bronchiectasis.
stress. Thorax 53, 867–870 (1998). 67–73 (2013). Cochrane Database Syst. Rev. 3, CD012406 (2018).
105. Loukides, S., Horvath, I., Wodehouse, T., Cole, P. J. 129. Chang, A. B., Boyce, N. C., Masters, I. B., Torzillo, P. J. 153. Serisier, D. J. et al. Effect of long-​term, low-​dose
& Barnes, P. J. Elevated levels of expired breath & Masel, J. P. Bronchoscopic findings in children with erythromycin on pulmonary exacerbations among
hydrogen peroxide in bronchiectasis. Am. J. Respir. non-​cystic fibrosis chronic suppurative lung disease. patients with non-​cystic fibrosis bronchiectasis: the
Crit. Care Med. 158, 991–994 (1998). Thorax 57, 935–938 (2002). BLESS randomized controlled trial. JAMA 309,
106. Willis, D., Moore, A. R., Frederick, R. & Willoughby, D. A. 130. Douros, K. et al. Bronchoscopic and high-​resolution CT 1260–1267 (2013).
Heme oxygenase: a novel target for the modulation of scan findings in children with chronic wet cough. Chest 154. Wong, C. et al. Azithromycin for prevention of
the inflammatory response. Nat. Med. 2, 87–90 (1996). 140, 317–323 (2011). exacerbations in non-​cystic fibrosis bronchiectasis
107. Chalmers, J. D., McHugh, B. J., Docherty, C., 131. Martinez-​Garcia, M. A. et al. Multidimensional (EMBRACE): a randomised, double-​blind,
Govan, J. R. W. & Hill, A. T. Vitamin-​D deficiency is approach to non-​cystic fibrosis bronchiectasis: the placebo-​controlled trial. Lancet 380, 660–667
associated with chronic bacterial colonisation and FACED score. Eur. Respir. J. 43, 1357–1367 (2014). (2012).
disease severity in bronchiectasis. Thorax 68, 39–47 132. Ellis, H. C., Cowman, S., Fernandes, M., Wilson, R. & The first-​published of three important macrolide
(2013). Loebinger, M. R. Predicting mortality in bronchiectasis studies conducted in adults, demonstrating
108. Chang, A. B. et al. Chronic suppurative lung disease and using bronchiectasis severity index and FACED scores: consistent reductions in exacerbation frequency
bronchiectasis in children and adults in Australia a 19-year cohort study. Eur. Respir. J. 47, 482–489 with long-​term low-​dose macrolide versus placebo.
and New Zealand Thoracic Society of Australia and (2016). 155. Altenburg, J. et al. Effect of azithromycin maintenance
New Zealand guidelines. Med. J. Aust. 202, 21–23 133. Mao, B., Yang, J.-W., Lu, H.-W. & Xu, J.-F. Asthma and treatment on infectious exacerbations among patients
(2015). risk of bronchiectasis exacerbation: we still need more with non-​cystic fibrosis bronchiectasis: the BAT
109. Dimakou, K. et al. Non CF-​bronchiectasis: aetiologic evidence. Eur. Respir. J. 48, 1247–1248 (2016). randomized controlled trial. JAMA 309, 1251–1259
approach, clinical, radiological, microbiological and 134. Araujo, D. et al. Standardised classification of the (2013).
functional profile in 277 patients. Respir. Med. 116, aetiology of bronchiectasis using an objective 156. Valery, P. C. et al. Long-​term azithromycin for
1–7 (2016). algorithm. Eur. Respir. J. 50, 1701289 (2017). Indigenous children with non-​cystic-fibrosis
110. McCallum, G. B. & Binks, M. J. The epidemiology of 135. Gaillard, E. A., Carty, H., Heaf, D. & Smyth, R. L. bronchiectasis or chronic suppurative lung disease
chronic suppurative lung disease and bronchiectasis in Reversible bronchial dilatation in children: comparison (Bronchiectasis Intervention Study): a multicentre,
children and adolescents. Front. Pediatr. 5, 27 (2017). of serial high-​resolution computer tomography scans of double-​blind, randomised controlled trial. Lancet
111. Kapur, N., Masters, I. B. & Chang, A. B. Exacerbations the lungs. Eur. J. Radiol. 47, 215–220 (2003). Respir. Med. 1, 610–620 (2013).
in noncystic fibrosis bronchiectasis: clinical features 136. McCallum, G. B. & Chang, A. B. “Good enough” is A multicentre randomized controlled trial
and investigations. Respir. Med. 103, 1681–1687 “not enough” when managing indigenous adults with demonstrating reduced exacerbations in
(2009). bronchiectasis in Australia and New Zealand. Indigenous children treated with azithromycin
112. Chang, A. B., Redding, G. J. & Everard, M. L. Chronic Respirology 23, 725–726 (2018). compared with placebo.
wet cough: protracted bronchitis, chronic suppurative 137. Aliberti, S. et al. Quality standards for the 157. van Ingen, J. et al. Poor adherence to management
lung disease and bronchiectasis. Pediatr. Pulmonol. management of bronchiectasis in Italy: a national guidelines in nontuberculous mycobacterial pulmonary
43, 519–531 (2008). audit. Eur. Respir. J. 48, 244–248 (2016). diseases. Eur. Respir. J. 49, 1601855 (2017).
113. Goyal, V., Grimwood, K., Marchant, J., Masters, I. B. 138. Kwak, H. J. et al. High prevalence of bronchiectasis 158. Haworth, C. S., Foweraker, J. E., Wilkinson, P.,
& Chang, A. B. Does failed chronic wet cough response in adults: analysis of CT findings in a health screening Kenyon, R. F. & Bilton, D. Inhaled colistin in patients
to antibiotics predict bronchiectasis? Arch. Dis. Child program. Tohoku J. Exp. Med. 222, 237–242 with bronchiectasis and chronic Pseudomonas
99, 522–525 (2014). (2010). aeruginosa infection. Am. J. Respir. Crit. Care Med.
114. Suarez-​Cuartin, G., Chalmers, J. D. & Sibila, O. 139. Flude, L. J., Agent, P. & Bilton, D. Chest physiotherapy 189, 975–982 (2014).
Diagnostic challenges of bronchiectasis. Respir. Med. techniques in bronchiectasis. Clin. Chest Med. 33, 159. Barker, A. F. et al. Aztreonam for inhalation solution
116, 70–77 (2016). 351–361 (2012). in patients with non-​cystic fibrosis bronchiectasis
115. van der Bruggen-​Bogaarts, B. A., van der Bruggen, H. M., 140. Munoz, G., de Gracia, J., Buxo, M., Alvarez, A. & (AIR-BX1 and AIR-​BX2): two randomised double-​blind,
van Waes, P. F. & Lammers, J. W. Assessment of Vendrell, M. Long-​term benefits of airway clearance in placebo-​controlled phase 3 trials. Lancet Respir. Med.
bronchiectasis: comparison of HRCT and spiral volumetric bronchiectasis: a randomised placebo-​controlled trial. 2, 738–749 (2014).
CT. J. Comput. Assist. Tomogr. 20, 15–19 (1996). Eur. Respir. J. 51, 1701926 (2018). At the time, the largest inhaled antibiotic trial in
116. Pasteur, M. C., Bilton, D. & Hill, A. T. British Thoracic 141. Wong, C., Sullivan, C. & Jayaram, L. ELTGOL airway bronchiectasis; this study found that aztreonam,
Society guideline for non-​CF bronchiectasis. Thorax clearance in bronchiectasis: laying the bricks of which is licensed for use in cystic fibrosis, did not
65, 577 (2010). evidence. Eur. Respir. J. 51, 1702232 (2018). improve QOL in bronchiectasis and was associated
117. Matsuoka, S. et al. Bronchoarterial ratio and bronchial 142. Lee, A. L., Burge, A. T. & Holland, A. E. Airway with increased adverse events.
wall thickness on high-​resolution CT in asymptomatic clearance techniques for bronchiectasis. Cochrane 160. De Soyza, A. et al. RESPIRE 1: a phase III placebo-​
subjects: correlation with age and smoking. AJR Am. J. Database Syst. Rev. 11, CD008351 (2015). controlled randomised trial of ciprofloxacin dry
Roentgenol. 180, 513–518 (2003). 143. Nicolson, C. H. H. et al. The long term effect of powder for inhalation in non-​cystic fibrosis
118. Berend, N., Woolcock, A. J. & Marlin, G. E. inhaled hypertonic saline 6% in non-​cystic fibrosis bronchiectasis. Eur. Respir. J. 51, 1702052 (2018).
Relationship between bronchial and arterial diameters bronchiectasis. Respir. Med. 106, 661–667 (2012). One of two studies describing the largest inhaled
in normal human lungs. Thorax 34, 354–358 (1979). 144. Lee, A. L., Hill, C. J., McDonald, C. F. & Holland, A. E. antibiotic development programme in
119. Diaz, A. A. et al. Bronchoarterial ratio in never-​ Pulmonary rehabilitation in individuals with non-​cystic bronchiectasis; this study found no consistent
smokers adults: implications for bronchial dilation fibrosis bronchiectasis: a systematic review. Arch. Phys. improvements in exacerbation frequency or QOL
definition. Respirology 22, 108–113 (2017). Med. Rehabil. 98, 774–782 (2017). with inhaled antibiotics, questioning their role as a
120. Kapur, N., Masel, J. P., Watson, D., Masters, I. B. & 145. Kuehni, C. E., Goutaki, M. & Kobbernagel, H. E. therapy for patients with bronchiectasis.
Chang, A. B. Bronchoarterial ratio on high-​resolution Hypertonic saline in patients with primary ciliary 161. Aksamit, T. et al. RESPIRE 2: a phase III
CT scan of the chest in children without pulmonary dyskinesia: on the road to evidence-​based treatment placebo-​controlled randomised trial of ciprofloxacin
pathology: need to redefine bronchial dilatation. Chest for a rare lung disease. Eur. Respir. J. 49, 1602514 dry powder for inhalation in non-​cystic fibrosis
139, 1445–1450 (2011). (2017). bronchiectasis. Eur. Respir. J. 51, 1702053 (2018).
121. Chalmers, J. D. Bronchiectasis and COPD overlap: 146. Bilton, D. et al. Inhaled mannitol for non-​cystic fibrosis 162. Chotirmall, S. H. & Chalmers, J. D. RESPIRE:
a case of mistaken identity? Chest 151, 1204–1206 bronchiectasis: a randomised, controlled trial. Thorax breathing new life into bronchiectasis. Eur. Respir. J.
(2017). 69, 1073–1079 (2014). 51, 1702444 (2018).
122. Kim, S. J. et al. Normal bronchial and pulmonary 147. Guan, W.-J., Huang, Y., Chen, C.-L., Chen, R.-C. & 163. Barker, A. F. et al. Tobramycin solution for inhalation
arterial diameters measured by thin section CT. Zhong, N.-S. Macrolides, mucoactive drugs and reduces sputum Pseudomonas aeruginosa density in
J. Comput. Assist. Tomogr. 19, 365–369 (1995). adherence for the management of bronchiectasis. bronchiectasis. Am. J. Respir. Crit. Care Med. 162,
123. Brody, A. & Chang, A. The imaging definition of Eur. Respir. J. 51, 1701987 (2018). 481–485 (2000).
bronchiectasis in children: is it time for a change? 148. Chalmers, J. D. & Polverino, E. Macrolides, mucoactive 164. Drobnic, M. E., Suñé, P., Montoro, J. B., Ferrer, A.
Pediatr. Pulmonol. 53, 6–7 (2018). drugs and adherence for the management of & Orriols, R. Inhaled tobramycin in non-​cystic
124. Hill, L. E., Ritchie, G., Wightman, A. J., Hill, A. T. & bronchiectasis. Eur. Respir. J. 51, 1702033 (2018). fibrosis patients with bronchiectasis and chronic
Murchison, J. T. Comparison between conventional 149. O’Donnell, A. E., Barker, A. F., Ilowite, J. S. & Fick, R. B. bronchial infection with Pseudomonas aeruginosa.
interrupted high-​resolution CT and volume Treatment of idiopathic bronchiectasis with Ann. Pharmacother. 39, 39–44 (2005).

Nature Reviews | Disease Primers | Article citation ID: (2018) 4:45 17

0123456789();
P­r i­m e­r

165. Vendrell, M., Muñoz, G. & de Gracia, J. Evidence of 186. Coulter, T. I. et al. Clinical spectrum and features of 207. Tan, H.-L. et al. The Th17 pathway in cystic fibrosis
inhaled tobramycin in non-​cystic fibrosis bronchiectasis. activated phosphoinositide 3-kinase delta syndrome: lung disease. Am. J. Respir. Crit. Care Med. 184,
Open Respir. Med. J. 9, 30–36 (2015). a large patient cohort study. J. Allergy Clin. Immunol. 252–258 (2011).
166. Brodt, A. M., Stovold, E. & Zhang, L. Inhaled antibiotics 139, 597–606 (2017). 208. Picard, C. et al. Primary immunodeficiency diseases:
for stable non-​cystic fibrosis bronchiectasis: a systematic 187. Goeminne, C. P. et al. The impact of acute air pollution an update on the classification from the International
review. Eur. Respir. J. 44, 382–393 (2014). fluctuations on bronchiectasis pulmonary exacerbation. Union of Immunological Societies Expert Committee
167. Murray, M. P. et al. A randomized controlled trial of A case-​crossover analysis. Eur. Respir. J. 52, 1702557 for Primary Immunodeficiency 2015. J. Clin. Immunol.
nebulized gentamicin in non-​cystic fibrosis (2018). 35, 696–726 (2015).
bronchiectasis. Am. J. Respir. Crit. Care Med. 183, 188. Chalmers, J. D. et al. Cross-​infection risk in patients 209. Zemanick, E. T. et al. Airway microbiota across age
491–499 (2011). with bronchiectasis: a position statement from the and disease spectrum in cystic fibrosis. Eur. Respir. J.
168. Haworth, C. et al. Inhaled liposomal ciprofloxacin in European Bronchiectasis Network (EMBARC), 50, 1700832 (2017).
patients with bronchiectasis and chronic Pseudomonas EMBARC/ELF patient advisory group and European 210. Paff, T. et al. A randomised controlled trial on the effect
aeruginosa infection: results from two parallel phase III Reference Network (ERN-​Lung) Bronchiectasis of inhaled hypertonic saline on quality of life in primary
trials (ORBIT-3 and -4) [abstract]. Am. J. Respir. Crit. Network. Eur. Respir. J. 51, 1701937 (2018). ciliary dyskinesia. Eur. Respir. J. 49, 1601770 (2017).
Care Med. 195, A7604 (2018). 189. Blasi, F., Chalmers, J. D. & Aliberti, S. COPD and 211. Gupta, S. et al. Qualitative analysis of high-​resolution
169. Contoli, M. et al. Long-​term effects of inhaled bronchiectasis: phenotype, endotype or co-​morbidity? CT scans in severe asthma. Chest 136, 1521–1528
corticosteroids on sputum bacterial and viral loads in COPD 11, 603–604 (2014). (2009).
COPD. Eur. Respir. J. 50, 1700451 (2017). 190. Aliberti, S. et al. Clinical phenotypes in adult patients with 212. Agarwal, R. et al. A randomised trial of glucocorticoids
170. Kapur, N., Petsky, H. L., Bell, S., Kolbe, J. & Chang, A. B. bronchiectasis. Eur. Respir. J. 47, 1113–1122 (2016). in acute-​stage allergic bronchopulmonary aspergillosis
Inhaled corticosteroids for bronchiectasis. Cochrane 191. Pasteur, M. C. et al. An investigation into causative complicating asthma. Eur. Respir. J. 47, 490–498
Database Syst. Rev. 5, CD000996 (2018). factors in patients with bronchiectasis. Am. J. Respir. (2016).
171. De Soyza, A. et al. A randomised, placebo-​controlled Crit. Care Med. 162, 1277–1284 (2000). 213. McShane, P. J., Naureckas, E. T. & Strek, M. E.
study of the CXCR2 antagonist AZD5069 in 192. Agustí, A. et al. Precision medicine in airway diseases: Bronchiectasis in a diverse US population: effects of
bronchiectasis. Eur. Respir. J. 46, 1021–1032 (2015). moving to clinical practice. Eur. Respir. J. 50, ethnicity on etiology and sputum culture. Chest 142,
172. Stockley, R. et al. Phase II study of a neutrophil 1701655 (2017). 159–167 (2012).
elastase inhibitor (AZD9668) in patients with 193. Agusti, A. et al. Treatable traits: toward precision A detailed description of a US-​based cohort that
bronchiectasis. Respir. Med. 107, 524–533 (2013). medicine of chronic airway diseases. Eur. Respir. J. 47, reveals differences in aetiology and microbiology
173. Bedi, P. et al. A randomized controlled trial of 410–419 (2016). based on ethnicity, a finding that has become
atorvastatin in patients with bronchiectasis infected 194. Vedel-​Krogh, S., Nordestgaard, B. G., Lange, P., increasingly important as more global data on
with Pseudomonas aeruginosa: a proof of concept Vestbo, J. & Nielsen, S. F. Blood eosinophil count and bronchiectasis become available.
study. Chest 152, 368–378 (2017). risk of pneumonia hospitalisations in individuals with
174. Mandal, P. et al. Atorvastatin as a stable treatment in COPD. Eur. Respir. J. 51, 1800120 (2018). Acknowledgements
bronchiectasis: a randomised controlled trial. Lancet 195. Kerkhof, M. et al. Blood eosinophil count and J.D.C. is supported by the GSK/British Lung Foundation Chair
Respir. Med. 2, 455–463 (2014). exacerbation risk in patients with COPD. Eur. Respir. of Respiratory Research. A.B.C. is supported by an Australian
175. Konstan, M. W. et al. A randomized double blind, J. 50, 1700761 (2017). National Health and Medical Research Council Practitioner
placebo controlled phase 2 trial of BIIL 284 BS (an LTB4 196. Southworth, T., Beech, G., Foden, P., Kolsum, U. & Fellowship (grant 105821). S.H.C. is supported by the
receptor antagonist) for the treatment of lung disease in Singh, D. The reproducibility of COPD blood eosinophil Singapore Ministry of Health’s National Medical Research
children and adults with cystic fibrosis. J. Cyst. Fibros counts. Eur. Respir. J. 52, 1800427 (2018). Council under its Transition Award (NMRC/TA/0048/2016),
13, 148–155 (2014). 197. Palmer, R. et al. Dipeptidyl peptidase 1 inhibitor the Lee Kong Chian School of Medicine, Nanyang
176. Fan, L.-C., Liang, S., Lu, H.-W., Fei, K. & Xu, J.-F. AZD7986 induces a sustained, exposure-​dependent Technological University Start-​Up Grant and would like to
Efficiency and safety of surgical intervention to patients reduction in neutrophil elastase activity in healthy acknowledge the Academic Respiratory Initiative for
with non-​cystic fibrosis bronchiectasis: a meta-​analysis. subjects. Clin. Pharmacol. Ther. https://doi.org/10.1002/ Pulmonary Health (TARIPH).
Sci. Rep. 5, 17382 (2015). cpt.1053 (2018).
177. Birch, J. et al. Outcomes of lung transplantation in adults 198. Harding, R. & Maritz, G. Maternal and fetal origins of Author contributions
with bronchiectasis. BMC Pulm. Med. 18, 82 (2018). lung disease in adulthood. Semin. Fetal Neonatal Med. Introduction (J.D.C.); Epidemiology (P.J.M.); Mechanisms/
178. Hill, A. T. et al. Pulmonary exacerbation in adults with 17, 67–72 (2012). pathophysiology (S.H.C.); Diagnosis, screening and preven-
bronchiectasis: a consensus definition for clinical 199. Maritz, G. S. & Harding, R. Life-​long programming tion (A.B.C.); Management (R.D.); Quality of life (P.J.M.);
research. Eur. Respir. J. 49, 1700051 (2017). implications of exposure to tobacco smoking and Outlook (J.D.C.); Overview of Primer (J.D.C.).
A global consensus document on the definition of nicotine before and soon after birth: evidence for
exacerbation for use in clinical research; this paper altered lung development. Int. J. Environ. Res. Public Competing interests
is notable for the involvement of investigators from Health 8, 875–898 (2011). J.D.C. has been an investigator, advisory board member or
Europe, North America, Australasia and Africa in a 200. Caskey, S. et al. Structural and functional lung trial steering committee member for several bronchiectasis
global task force. impairment in adult survivors of bronchopulmonary clinical trials, including for Aradigm, Bayer, Grifols, Novartis
179. O’Leary, C. J. et al. Relationship between psychological dysplasia. Ann. Am. Thorac Soc. 13, 1262–1270 and Zambon. He is chair of the European Bronchiectasis
well-​being and lung health status in patients with (2016). Registry. A.B.C. is a member of the data safety monitoring
bronchiectasis. Respir. Med. 96, 686–692 (2002). 201. Valery, P. C. et al. Hospital-​based case–control study board for an unlicensed vaccine study (for GlaxoSmithKline)
180. Ryu, Y. J., Chun, E.-M., Lee, J. H. & Chang, J. H. of bronchiectasis in Indigenous children in central and an adviser for study design of an unlicensed product for
Prevalence of depression and anxiety in outpatients Australia. Pediatr. Infect. Dis. J. 23, 902–908 (2004). cough (for Merck). She declares no financial conflicts of inter-
with chronic airway lung disease. Korean J. Intern. 202. Jayes, L. et al. SmokeHaz: systematic reviews and est regarding the content of this manuscript. P.J.M. has
Med. 25, 51–57 (2010). meta-​analyses of the effects of smoking on respiratory served on the advisory committee to the FDA for Bayer and
181. Joish, V. N., Spilsbury-​Cantalupo, M., Operschall, E., health. Chest 150, 164–179 (2016). is an advisory board member for Aradigm, Bayer, Grifols, Hill
Luong, B. & Boklage, S. Economic burden of non-​cystic 203. Walker, C. L. F. et al. Global burden of childhood Rom and Insmed. She declares no financial conflicts of inter-
fibrosis bronchiectasis in the first year after diagnosis pneumonia and diarrhoea. Lancet 381, 1405–1416 est regarding the content of this manuscript. S.H.C. and R.D.
from a US health plan perspective. Appl. Health Econ. (2013). declare no competing interests.
Health Policy 11, 299–304 (2013). 204. Chang, A. B. et al. Protracted bacterial bronchitis:
182. Kapur, N., Masters, I. B., Newcombe, P. & Chang, A. B. the last decade and the road ahead. Pediatr. Pulmonol. Publisher’s note
The burden of disease in pediatric non-​cystic fibrosis 51, 225–242 (2016). Springer Nature remains neutral with regard to jurisdictional
bronchiectasis. Chest 141, 1018–1024 (2012). 205. Sjogren, P., Nilsson, E., Forsell, M., Johansson, O. & claims in published maps and institutional affiliations.
183. Murray, M. P., Turnbull, K., MacQuarrie, S., Hoogstraate, J. A systematic review of the preventive
Pentland, J. L. & Hill, A. T. Validation of the Leicester effect of oral hygiene on pneumonia and respiratory Reviewer information
cough questionnaire in non-​cystic fibrosis tract infection in elderly people in hospitals and Nature Reviews Disease Primers thank L.-A. Daniel, K. Olivier,
bronchiectasis. Eur. Respir. J. 34, 125–131 (2009). nursing homes: effect estimates and methodological E. Polverino, H. Tiddens and the other anonymous reviewer(s)
184. Guan, W.-J. et al. Inflammatory responses, spirometry, quality of randomized controlled trials. J. Am. Geriatr. for their contribution to the peer review of this work.
and quality of life in subjects with bronchiectasis Soc. 56, 2124–2130 (2008).
exacerbations. Respir. Care 60, 1180–1189 (2015). 206. Singleton, R. J. et al. Indigenous children from three Related links
185. Courtney, J. M. et al. Quality of life and inflammation countries with non-​cystic fibrosis chronic suppurative European Multicentre Bronchiectasis Audit and Research
in exacerbations of bronchiectasis. Chron. Respir. Dis. lung disease/bronchiectasis. Pediatr. Pulmonol. 49, Collaboration (EMBARC): www.bronchiectasis.eu
5, 161–168 (2008). 189–200 (2014).

18 | Article citation ID: (2018) 4:45 www.nature.com/nrdp

0123456789();

You might also like