You are on page 1of 13

The n e w e ng l a n d j o u r na l of m e dic i n e

Review Article

Jeffrey M. Drazen, M.D., Editor

Muco-Obstructive Lung Diseases


Richard C. Boucher, M.D.​​

A 
spectrum of lung diseases that affect the airways, including From the Marsico Lung Institute, University
chronic obstructive pulmonary disease (COPD), cystic fibrosis, primary of North Carolina at Chapel Hill, Chapel Hill.
Address reprint requests to Dr. Boucher
ciliary dyskinesia, and non–cystic fibrosis bronchiectasis, can be character- at the Marsico Lung Institute, University
ized as muco-obstructive diseases.1-4 These diseases have the clinical features of of North Carolina at Chapel Hill, 7008
cough, sputum production, and episodic exacerbations that are often associated Marsico Hall, Chapel Hill, NC 27599, or at
­richard_boucher@​­med​.­unc​.­edu.
with a diagnosis of chronic bronchitis.5 However, neither “chronic bronchitis” nor
“hypersecretory diseases”6 adequately describes the diffuse mucus obstruction, N Engl J Med 2019;380:1941-53.
DOI: 10.1056/NEJMra1813799
airway-wall ectasia, chronic inflammation, and bacterial infection that are typical Copyright © 2019 Massachusetts Medical Society.
of these conditions; therefore, “muco-obstructive” may be a preferred descriptive
term. Although asthma can also be associated with diffuse airway mucus obstruc-
tion,7 its distinct pathophysiological mechanisms preclude discussion in this
grouping.8
The pathogenesis of muco-obstruction of the airways is depicted in Figure 1A.
In healthy persons, a well-hydrated mucus layer is transported rapidly (at a rate of
approximately 50 μm per second) from the distal airways toward the trachea. In
muco-obstructive diseases, epithelial defects in ion–fluid transport, mucin secre-
tion, or a combination of these lead to hyperconcentrated (dehydrated) mucus,
failed mucus transport, and mucus adhesion to airway surfaces. Mucus that is
accumulated in the trachea can be expectorated by cough as phlegm or sputum.11
Mucus in the small airways cannot be cleared by cough and accumulates, forming
the nidus for airflow obstruction, infection, and inflammation.11

Bio chemic a l a nd Bioph ysic a l Proper t ie s of Mucus


R el at i v e t o A irwa y F unc t ion

Human airway mucus is a hydrogel composed of approximately 98% water, 0.9%


salt, 0.8% globular proteins, and 0.3% high-molecular-weight mucin polymers.12
The hydration (concentration) status of mucus is measured as the wet-to-dry con-
tent of mucus (i.e., the percentage of a given volume that consists of nonvolatile
solids) or as the mucin concentration determined by light-scattering tech-
niques.9,11,13 The correlation between the two measurements (in healthy persons or
those with muco-obstructive disease) is high, which allows both measures to be
used to describe this mucus property.9
The two major synthesized and secreted respiratory mucins, MUC5B and
MUC5AC, are physically very large, spanning 0.2 to 10 μm in length for single
polymers. The secreted mucin polymers interweave to form mesh-like gels with
mesh sizes that are concentration dependent (i.e., higher concentrations are associ-
ated with smaller mesh sizes). MUC5B and MUC5AC share many features, includ-
ing their multimeric organization and high carbohydrate content (approximate-
ly 75% of total weight) (Fig. 1B).14 There are, however, major differences in the

n engl j med 380;20 nejm.org  May 16, 2019 1941


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Disease Progression

Normal Mucus stasis Cough


Muco-obstructive lung disease
Airflow Mucin

Cl –

MCC=0 µm/sec
MCC=50 µm/sec

M UCUS
LA YE R
Mucin
Mucin
PCL Cl– H 2O Na + H 2O
Cl –
Accumulation
ENaC CACC
and
CFTR Obstruction and infection

Mucin
Mucin
Mucin
Cl –

Na+ H 2O Na + H 2O Na + H 2O

B Molecular Domain Structures and Relative Sizes of Mucins


Molecular Mass Protein Length and Structure
>4 µm

400 kDa MUC5AC

400 kDa MUC5B

66 kDa ALB
Glycosylated region
200 kDa MUC1 250 nm

900 kDa MUC4 1 µm

3500 kDa MUC16 4 µm

C Total Sputum Mucin Concentrations

8000
Mucin Concentration (µg/ml)

6000

4000

2000

0
Healthy Persons Patients with Patients with Patients with Patients with
(N=69) COPD (N=359) NCFB (N=99) CF (N=20) PCD (N=42)

1942 n engl j med 380;20 nejm.org May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

Figure 1 (facing page). Muco-Obstructive Disease


dominant mucin (10:1 over MUC5AC) in normal
Pathogenesis, Mucin Species, and Total Mucin human lower airways.10
­Concentrations in Health and Disease. The physical properties of mucus can be pre-
Panel A shows the progression from normal to muco- dicted from polymer physics15 that describe not
obstructed airways. In healthy persons (left), well-­ only well-studied mucus viscoelastic properties
balanced epithelial sodium (Na+) absorption and secre- but also other key biophysical properties, includ-
tion of chloride anions (Cl −) hydrates airway surfaces
and promotes efficient mucociliary clearance (MCC).
ing mucus osmotic pressure, adhesion, cohesion,
In persons with muco-obstructive lung disease (middle), and friction.11,13,15 The viscoelastic and osmotic
an imbalance of ion transport coupled with mucin hyper- properties of mucus, as with other polymer gels,
secretion increases mucin concentrations in the mucus scale to the third power, or higher, of concentra-
layer, osmotically compresses the periciliary layer (PCL), tion. This feature of mucus accounts for the
and abolishes MCC. The adherent mucus may be ex-
pelled as sputum by cough (upper right). Mucus that
observation that relatively small changes in con-
cannot be expelled by cough accumulates, concentrates, centration have profound effects on the bio-
obstructs airflow, and becomes the nidus for infection physical and transport properties of mucus —
(lower right). CFTR denotes cystic fibrosis transmem- for example, higher concentrations dramatically
brane conductance regulator, and ENaC the epithelial decrease transportability.13,16 In addition, the
sodium channel. Panel B shows domain structures and
relative sizes of the secreted mucins (MUC5AC and
properties of mucus gels can be modified by
MUC5B) and tethered mucins (MUC1, MUC4, and conditions that create inter-mucin polymer in-
MUC16). For reference, the globular protein albumin teractions with relatively long bond half-lives,
(ALB) is shown. The secreted mucins are composed of producing “sticky” polymer gels whose viscous
monomers with N terminals (MUC5AC, green; MUC5B, properties can scale to the eighth power of con-
light blue), glycosylated domains, and C terminals
(MUC5AC, yellow; MUC5B, purple). Both the C–C ter-
centration.13,15
minal dimers and N–N terminal multimers are linked
by S–S bonds. The glycosylated domains contain sugar
side chains from 2 to 15 sugar molecules in length, often
Mucin C oncen t r at ions in Muc o -
terminally capped with sialic acid or sulfate, which gives
Obs t ruc t i v e Lung Dise a se s
mucins a negative charge. The glycosylation domains
provide mucin hydration, reflecting the avidity of sugar
Mucin concentrations are abnormally raised in
molecules for water, and a combinatorial library of bind- each of the four muco-obstructive diseases —
ing sites capable of trapping most inhaled materials COPD, cystic fibrosis, primary ciliary dyskinesia,
with a low but sufficient binding affinity to mediate and non–cystic fibrosis bronchiectasis — and
clearance. The tethered mucins have cytoplasmic these findings are consistent with a role for ele-
N-terminal (dark gray), transmembrane (light gray),
and heavily glycosylated extracellular domains. For sim-
vated mucin concentrations in the pathogenesis
plicity, unique domains and proteolytic cleavage sites of muco-obstructive disease (Fig. 1C). Although
are not shown. Panel C shows total sputum mucin MUC5AC levels are elevated and may uniquely
concentrations in healthy persons and patients with modify mucus properties, MUC5B is the domi-
chronic obstructive pulmonary disease (COPD), non– nant mucin by concentration in these diseases.10
cystic fibrosis bronchiectasis (NCFB), cystic fibrosis
(CF), or primary ciliary dyskinesia (PCD). Sputum mucin
concentrations were measured by means of high-per- In tegr ated Cil i a a nd C ough-
formance liquid chromatography and refractometry.9,10 Dependen t Mucus Cl e a r a nce
T bars indicate standard errors of the mean.
The classic views of the mucus transport system
have undergone substantial revision at both
regulation of transcription and probably the macroscopic and microscopic length scales dur-
function of MUC5B and MUC5AC. Data from ing the past decade.13,17
studies in mice indicate that Muc5b is required
for basal mucociliary transport, whereas Muc5ac Macroscopic — Intrapulmonary Mucin
reacts to external stresses, features that are Sources
likely to be mimicked in humans. Recent data Mucus lines all airway surfaces as it moves to-
indicate that MUC5B is by concentration the ward the throat through ciliary activity. Tradi-

n engl j med 380;20 nejm.org  May 16, 2019 1943


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Models of Mucus Transport B Normal Epithelial Ion and Water Transport


Gel on liquid Two gels MCC = 50 µm/sec
M U C U S LA YER M UC US L A Y E R MU C U S L A Y E R
πML = 100 Pa 2% solids
MUC5AC MUC5AC
PCL Cl –
MUC5B MUC5B πPCL = 500 Pa ATP AD O Cl – H 2O

A2b
ENaC P2Y2
SOL PCL
MUC16 CaCC CFTR

MUC4
Mucin

MUC1

E P I T H E LIA L C ELL Na + H 2O

C Water-Drawing Power of the Mucus Layer (πML) vs. the PCL (πPCL) D Mucus Concentration and Osmotic Pressure
Normal steady state πML<πPCL Diseased hyperabsorption πML=πPCL
10,000
πML Normal
CF Sputum
CF Lung
Osmotic Pressure (Pa)

1000

πPCL
πPCL
WATER
W A TER 100

10
0
0.8 1 2 3 4 6 8 10 14
Mucus Concentration (% solids)
E P I T H E LIA L C ELL

E Mucus-Layer Concentration vs. Predicted Clearance F Airflow, Mucus, and Cough Efficiency

πML < πPCL


Relative Mucus Clearance (%)

πML ≈ πPCL
100 Adhesion
80 Cohesion
A I RFL O W

60
πML > πPCL
40
20 Viscosity
Friction
0
0 2 4 6 8 10 12
% Solids

100 98 96 94 92 90 88
% Water

1944 n engl j med 380;20 nejm.org May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

Figure 2 (facing page). Two-Gel Model of Mucus Transport.


tionally, the superficial epithelia that line large
Panel A, left, shows the classic “gel-on-liquid” model show- (bronchial) and small (bronchiolar) airways were
ing a mucus layer (MUC5AC and MUC5B) and a “sol” layer believed to secrete MUC5AC, whereas large-air-
as liquid surrounding cilia. Panel A, right, shows the “two- way submucosal glands were thought to secrete
gel” model in which the mucus layer remains the same,
but a periciliary layer (gel) is formed by tethered macro-
MUC5B. Data from mouse models and human
molecules, including MUC1, MUC4, and MUC16. Panel B studies have led to a reformulation of this
shows epithelial ion and water transport in normal human view.18,19 Under baseline conditions, the superfi-
airway epithelia. The percentage of solids in the normal cial epithelia of both human large and small
mucus layer and osmotic pressures (π) of the normal mu-
cus layer (πML) and PCL (πPCL) in pascals (Pa) are noted. airways secrete MUC5B. Airway MUC5B secre-
An ENaC in the apical membrane mediates sodium and tion is supplemented by MUC5B secretion from
liquid absorption.21 In parallel, the epithelium secretes the glands that may be in part tonic but probably
chloride and bicarbonate anions through CFTR and calcium-
activated chloride channels (CaCC).21 The balance between
can be greatly increased with cough.20 A variety
active sodium absorption and secretion of chloride and bi- of pulmonary stresses trigger superficial epithe-
carbonate anions, and hence fluid flow, is regulated in part lial MUC5AC expression.12 Recent research sug-
by lumenal concentrations of extracellular ATP, interacting gests that MUC5AC secretion is superimposed
through P2Y2 receptors, and adenosine (ADO), interacting
through A2b receptors.22 Panel C describes the relative water- on superficial airway secretory (club) cells that
drawing powers of the mucus layer (πML) and PCL (πPCL) are competent for basal MUC5B secretion.18
interfaced to epithelial cell–mediated fluid absorption or
secretion (blue arrows). The length of the Hookean springs
Microscopic — Mucus Layer–Cilia Topography
(to the right of the blue arrows) denotes the height of the
PCL (purple) or mucus layer (green), and the spring diameter The classic description of mucociliary clearance
is inversely proportional to osmotic pressure. In a normal envisioned the movement of a mucus (gel) layer
state (left), πML is lower than πPCL, represented by a green
over a periciliary watery (liquid) layer, also called
spring (πML) with a diameter larger than the purple spring
(πPCL). In a dehydrated state (right), persistent or abnormal the “sol” layer.17 This description has been re-
absorption initially removes fluid from the lower-osmotic- placed by one depicting the mucociliary appara-
pressure mucus layer but ultimately coordinately removes tus as comprising two gels: a mucus layer and a
fluid from both the mucus layer and the PCL. The osmotic
pressures of both layers are increased and equalized (small- periciliary layer (Fig. 2A).13 The periciliary layer
er spring diameters) and volume depleted (springs short- was identified as a dense gel composed of gly-
ened). This state osmotically compresses the cilia and pro- copolymers, including the MUC1, MUC4, and
duces mucus stasis. Panel D shows the relationship between
MUC16 mucins, tethered to epithelial-cell sur-
mucus concentration (i.e., percent solids) and osmotic
pressure (Pa).9 Samples of mucus from healthy persons, faces and cilia. An important concept that
sputum from patients with cystic fibrosis, and mucus ob- emerged from this formulation is not only that
tained from excised lungs of patients with cystic fibrosis the mucociliary-clearance apparatus is composed
are shown. Osmotic pressures in the mucus layer that are
higher than the basal osmotic pressure of the PCL (black of two polymer hydrogels but also that they
dashed line) are predicted to produce collapse of the PCL compete for hydration. The force that mediates
and slow MCC.13 Panel E shows MCC as a function of polymer-gel hydration — that is, the water-
mucus-layer concentration predicted from the ratio of πML
to πPCL. At normal mucus hydration (2% solids and 98%
drawing power — is described as the osmotic
water), πML is below basal πPCL values, the PCL is well pressure of a hydrogel. Experimental measure-
hydrated, and mucus transport is efficient. With modest ments revealed that the osmotic pressure of the
mucus dehydration (3 to 4% solids and 96 to 97% water), normal mucus layer consisting of 2% solids is
πML slightly exceeds πPCL, modest compression of the
PCL results, and MCC slows. When mucus dehydration is approximately 100 Pa,9,13 whereas the periciliary
severe (7 to 10% solids and 90 to 93% water), the mucus layer is more concentrated, with an osmotic
layer osmotically compresses cilia, producing mucus stasis. pressure of approximately 500 Pa (Fig. 2B).13 The
Panel F shows interactions of airflow with mucus biophysical
properties relevant to cough efficiency. In more distal air-
higher osmotic pressure of the periciliary layer
ways (below bifurcation), reduced airflows (smaller arrows) in healthy persons ensures that it is well hydrated,
can only slide mucus along airway surfaces, a movement providing appropriate lubrication for ciliary activ-
limited by friction between mucus and cell surfaces. At air- ity and transport of the overlying mucus layer.13
way bifurcations, mucus remodels as it enters the proximal
airway, which has a reduced aggregate surface area, a pro-
cess limited by mucus viscosity.11 In large airways (e.g., the Integration of Epithelial Ion Transport
trachea), the higher airflow (larger arrow) fractures intra- and Mucus Hydration
mucus cohesive forces, adhesive forces between mucus
and the cell surface, or both to expel mucus as sputum.23
The two-gel model allows for quantitative pre-
dictions with respect to interactions between

n engl j med 380;20 nejm.org  May 16, 2019 1945


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

A Muco-Obstructed Lung before Inflammation B Insult-Triggered Formation of Mucus Plaque

Inactive Insult Activated


macrophage (virus or aspiration) macrophage

MCC=0 µm/sec
MCC=50 µm/sec

Mucin Mucin

+
E P I T HE L I A L C E L L S Mucin
Fluid Poor
hydration production

C Muco-Inflammatory Positive-Feedback Cycle


MCC=0 µm/sec

Interleukin-1β
+ Mucin + PO2

+ +
+ Hypoxia + Interleukin-1α

Fluid Poor PO2 +


hydration
Mucin
+ SPDEF + ERN2 + production + ERN2 + SPDEF +
IL1R1 Interleukin-8, CXCL1

Parallel neutrophil-
mediated inflammation

Figure 3. Muco-Inflammatory Positive-Feedback (“Vicious”) Cycles in Muco-Obstructive Disease.


Panel A shows the basal muco-obstructive disease state in which surface hydration and mucus concentration may be
relatively normal but vulnerable to muco-obstruction with insults (e.g., aspiration or viral infection). Panel B shows
insult-triggered formation of abnormal mucus plaque. Insults increase mucin secretion not accompanied by adequate
hydration owing to abnormal fluid-secretory responses. Hyperconcentrated mucus activates resident macrophages
(pink) and produces hypoxia in subjacent airway epithelia (blue). Panel C shows the generation of persistent, muco-
inflammatory positive-feedback cycles. Activated resident macrophages release interleukin-1β (left), and hypoxic
necrotic epithelia (blue) release interleukin-1α (right). Interleukin-1α and -1β activate epithelial interleukin-1 receptors
(IL1R1) to induce mucin biosynthesis mediated by SPDEF and ERN2 and expression of proinflammatory cytokines
and chemokines (e.g., IL8 and CXCL1). Accelerated mucin secretion without proper hydration worsens mucus hyper-
concentration on airway surfaces and stimulates hypoxic macrophages and epithelia to release additional interleukin-
1α and -1β in positive-feedback loops (denoted by a plus sign). Secretion of proinflammatory mediators induces
parallel neutrophil-mediated inflammation; parallel neutrophil-mediated, protease-induced mucin secretion; and other
positive-feedback cycles (not shown). PO2 denotes partial pressure of oxygen.

1946 n engl j med 380;20 nejm.org May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

mucus-layer concentration (hydration) and the Patho gene sis


efficiency of mucociliary transport. These pre- of Muc o - Obs t ruc t ion
dictions are best understood from analyses of
how the mucus and periciliary layers respond An important feature of muco-obstructive lung
to fluid transport by the underlying airway epi- diseases is disease heterogeneity — that is, there
thelium. are regions of the lung that are normal and
In healthy persons, airway epithelia can se- other regions within the same lung that are se-
crete or absorb ions and water and probably do verely diseased. Bronchiectasis is often a promi-
both simultaneously.21 Maintenance of normal nent finding in computed tomography (CT) of
airway-surface hydration is provided by cilial the lung. However, a common feature that uni-
mechanosensing of mucus-layer concentration, fies muco-obstructive diseases is the early mani-
regulated release of ATP by ciliated cells onto festation of disease in small airways (bronchioles),
the airway surface, and fine-tuning of ion-trans- as evidenced by pathological examination, micro-
port and fluid-transport rates through purino- computed tomography, and pulmonary-function
ceptor activation (Fig. 2B).8,24 In the well-hydrated studies.
normal state (2% solids and 98% water), the The airways are defended against infection
mucus layer acts as a fluid buffer to ensure ef- both by mechanical (mucus) clearance and by
ficient mucus transport (Fig. 2C; and Fig. S1 in antimicrobial proteins and peptides that are se-
the Supplementary Appendix, available with the creted into mucus by airway epithelial cells.25,26
text of this article at NEJM.org). However, in The ability of antimicrobial molecules and anti-
muco-obstructive disease, abnormal epithelial bodies to suppress bacterial-infection replication
fluid absorption depletes the airway surface of is short lived, probably on the order of hours.26
fluid, increases mucus concentrations, and raises Consequently, a kinetic “horse race” is manifest
osmotic pressures in the mucus layer to levels in the lung, pitting the rate of acquisition of
exceeding those in the periciliary layer (Fig. 2C). bacterial resistance to endogenous antimicrobial
Moderately hyperconcentrated mucus that is as- suppression against the rate of mechanical (mu-
sociated with disease compresses cilia and slows cus) bacterial clearance. However, slowing of
mucus transport, whereas severe hyperconcen- clearance alone is probably not sufficient to
tration flattens cilia and produces mucus stasis produce disease. It appears that the mucus layer
and adhesion. Thus, the two-gel formulation needs to come to a stop, with the formation of
predicts mucociliary transport rates from mucus mucus plaques and plugs within airway lumens,
concentrations (Fig. 2D and 2E) and explains to produce muco-obstructive disease. Studies in
why relatively small changes in hydration status animal models have shown that the full spec-
(2% vs. 8% solids [i.e., 98% vs. 92% water]) pro- trum of muco-obstructive diseases, including air-
duce disease. flow obstruction, inflammation, and intermittent
infection, are observed with mucus concentra-
Cough — Backup Mechanism Recruited tion–dependent formation of plaques and plugs
to Clear Adherent Mucus but not isolated defects in mucociliary clearance.27
When cilial-dependent mucus clearance fails, Mucus plaque or plug formation in the air-
cough becomes the backup clearance mecha- ways reflects an increase in mucin secretion,
nism.23 It is the combined failure of cilial-­ often stimulated by viral infections or aspira-
dependent and cough-dependent clearance that tion, coupled with poor epithelial hydration of
produces muco-obstructive disease. Like cilial- newly secreted mucins (Fig. 3A and 3B). On
dependent mucus transport, mucus concentra- formation of hyperconcentrated, static mucus
tion is a key determinant for all mucus proper- plaques, a bistable positive-feedback (“vicious”)
ties associated with cough-dependent transport, muco-inflammatory cycle is initiated (Fig. 3C).28,29
including friction, viscosity, cohesion, and adhe- Ultimately, severe or persistent muco-obstruc-
sion (Fig. 2F, and Fig. S2 in the Supplementary tion, typically associated with elevated airway
Appendix).11 levels of neutrophil elastase, drives the progres-

n engl j med 380;20 nejm.org  May 16, 2019 1947


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

sion from bronchitic to bronchiectatic or bron- muco-obstructive lung disease. Previous studies
chiolectatic lesions typical of these diseases.30,31 suggest that some exacerbations are caused by
Bacterial infection is a common feature of an intensification of disease in areas with pre-
muco-obstructive lung diseases.2-4,32 Data from existing disease, perhaps reflecting a change in
patients with cystic fibrosis or other muco-­ bacteria through genetic drift or phenotypic re-
obstructive diseases indicate that the major sites sponses to a local environment.40,41 However, the
of infection in muco-obstructive lung diseases new physical findings that are associated with
are intralumenal mucus plaques and plugs, not exacerbations suggest that many exacerbations
airway epithelial-cell surfaces.32 The limitation are associated with the spread of disease to pre-
of oxygen diffusion through mucus plaques to viously unaffected regions (Fig. S3 in the Supple-
underlying actively metabolizing epithelial cells mentary Appendix).42 A common mechanism for
produces regions of hypoxia within mucus spread may be gastric aspiration; this concept is
plaques (Fig. 3).32,33 Analyses of microbiome data consistent with data from patients with COPD,
from patients with cystic fibrosis or COPD have non–cystic fibrosis bronchiectasis, or cystic fi-
identified oral anaerobes as the first bacterial brosis.43,44 Perhaps the major trigger for spread
pathogens in the muco-obstructed lung.34,35 These is viruses aspirated from upper airways into the
data suggest that a mouth–lung aspiration axis, lung. A spread mechanism is consistent with
in which oral anaerobes infect hypoxic mucus data from patients with cystic fibrosis or non–
plaques, may be a unifying concept for early cystic fibrosis bronchiectasis showing that the
bacterial infection in most muco-obstructive dis- microbiome changes little during exacerba-
eases.35,36 Anaerobic bacteria can modify the tions.34,45 Clinically, exacerbations must be treat-
hypoxic mucus environment to promote invasion ed vigorously to limit permanent loss of lung
by classic pathogens (e.g., Pseudomonas aeruginosa), function.
which respond to mucus concentration–depen-
dent tightening of mucus meshes to grow as Dise a se-Specific Path wa ys
aggregates or biofilms.4,32,33,37,38 Future studies t o H y perc oncen t r at ion
are required to establish whether oral pharyn- of A irwa y Mucus
geal flora in sputum samples obtained from
patients with muco-obstructive disease may, like Cystic Fibrosis
classic pathogens, require treatment. Cystic fibrosis is the most extensively studied
disease with respect to the two-gel hypothesis
describing mucus concentration–dependent dis-
E x acer b at ions
ease. The abnormal concentration of mucus in
Exacerbations are typical of muco-obstructive the lungs of patients with cystic fibrosis reflects
diseases. An exacerbation is broadly defined as a primary abnormality of airway epithelial ion
a change in the patient’s perception of well- transport.4 In this disease, the airway epithelium
being, the seeking of health care, or a health is vulnerable to fluid hyperabsorption because of
care–implemented change in the patient’s medi- a defect in the secretion of chloride and bicar-
cal regimen.39 In all muco-obstructive lung dis- bonate anions mediated by the cystic fibrosis
eases, the overall rate of progression of disease transmembrane conductance regulator (CFTR) and
severity (e.g., loss of lung function) is probably by an intact sodium absorptive path. Laboratory
heavily influenced by the incidence and severity evidence suggests that cystic fibrosis can be trig-
of acute exacerbations. Common to all muco- gered by viral infections that lead to unre-
obstructive diseases, previous rates of exacer- strained liquid absorption, mucus hyperconcen-
bation and gastric aspiration are predictors of tration, and the formation of mucus plaques and
future rates of exacerbation — that is, a patient plugs (Fig. S4 in the Supplementary Appendix).22
with many past exacerbations is more likely to Mucus hyperconcentration as a feature of
have future exacerbations than a patient without cystic fibrosis was first documented in adults.
such a history.1 Sputum measurements revealed higher total
It is important to note that an exacerbation is mucin concentrations (Fig. 1C) and a higher
superimposed on preexisting, heterogeneous percentage of solids (Fig. S4 in the Supplemen-

1948 n engl j med 380;20 nejm.org  May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

tary Appendix) in patients with cystic fibrosis from patients with COPD, and higher mucus
than in healthy persons.9 Sputum osmotic pres- concentrations are correlated with reduced in
sures in healthy persons (approximately 100 Pa) vivo rates of mucociliary clearance.16 Data from
were below the osmotic pressure of the peri- a large cohort study involving patients with
ciliary layer (500 Pa), as predicted,13 whereas COPD suggest that a major trigger of COPD,
expectorated sputum samples from patients cigarette smoke, is associated with an increase
with cystic fibrosis had osmotic pressures slightly in mucin concentration.10 Furthermore, hyper-
above basal periciliary-layer values (≥500 Pa) concentration of mucins in sputum was associ-
(Fig. 2D). The mucus extracted from the lungs of ated in this cohort with disease severity, as in-
patients with cystic fibrosis at transplantation dexed by associations between increased mucin
had even higher concentrations (10 to 15% solids) concentrations and greater airflow obstruction
and osmotic pressures (>1000 Pa), findings con- and higher exacerbation rates.10 Increased mucin
sistent with a failure to clear (cough out) highly concentrations were also associated with a sub-
concentrated, adherent mucus.9,11 Optical coher- group of patients with early COPD (i.e., those
ence tomography of the upper airways in vivo of with nearly normal lung function), who were
adults with cystic fibrosis revealed evidence of shown to be at risk for rapid progression of their
mucus hyperconcentration, slowed mucociliary disease.10
clearance, and, as predicted by the two-gel hy- The airway epithelial defects that produce
pothesis, a reduction in the height of the peri- mucus hyperconcentration in COPD are com-
ciliary layer.46 plex.51 Exposure to cigarette smoke may produce
Recent data regarding bronchoalveolar lavage abnormalities in CFTR-mediated secretion of
from children with cystic fibrosis in the Austra- chloride anions through oxidant-induced reduc-
lian Respiratory Early Surveillance Team for tion of CFTR transcription rates and direct dam-
Cystic Fibrosis (AREST CF) cohort showed that a age to CFTR protein in the apical membrane.52,53
persistent increase in mucin concentration and Furthermore, increased extracellular nucleotide
inflammatory cells preceded bacterial infection and nucleoside metabolism decreases extracellu-
in the lung.47 “Rough” (hyperconcentrated) MUC5B lar ATP and adenosine levels, which suggests de-
and MUC5AC mucus flakes, probably of small- fective purinoceptor regulation of airway-surface
airway origin, were also a feature of bronchoal- hydration.16 These defects in epithelial ion and
veolar fluid from children with cystic fibrosis.48 fluid transport (hydration) are amplified by ciga-
Although failure of CFTR-dependent bicarbonate rette smoke–induced hypersecretion of MUC5AC
secretion might suggest a role for pH in early and MUC5B.6 Mucin hypersecretion may be par-
cystic fibrosis, transbronchoscopic measurements ticularly important in COPD because accelerated
in the AREST CF cohort did not detect a signifi- metabolism of co-released adenosine diphosphate,
cant difference in lower-airway pH between pa- adenosine monophosphate, and adenosine, cou-
tients with cystic fibrosis and disease controls pled with defective CFTR function, may limit
without cystic fibrosis. In addition, pH has less paracrine adenosine–CFTR hydration responses
effect than concentration on the biophysical that accompany mucin secretion.54
properties of mucus related to clearance or The mucin hyperconcentration that charac-
plaque formation.11 Aspirated oral anaerobes ap- terizes patients with COPD is perhaps the mild-
pear to be the first bacterial pathogens in the est example in the muco-obstructive disease
lungs of patients with cystic fibrosis, followed grouping (Fig. 1C). Patients with COPD also tend
by the classic gram-negative pathogens that to have the least severe muco-obstructive airway
probably accelerate the vicious cycle depicted in disease, particularly with respect to the lower
Figure 3 and loss of lung function.4,49,50 incidence of bronchiectasis and pseudomonas
infection (Fig. 2B).55,56
COPD
COPD manifests as persistent airflow obstruc- Primary Ciliary Dyskinesia
tion, most often in response to inhaled environ- The pathogenesis of primary ciliary dyskinesia
mental agents.1,51 Hyperconcentrated mucus has has typically been considered as solely a “motor
been reported in lower-airway samples obtained problem” caused by genetic abnormalities in

n engl j med 380;20 nejm.org  May 16, 2019 1949


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

cilial shaft proteins, cilial beat frequency, and with bronchiectasis as a final common path-
defective mucociliary clearance.3,57 However, way.60 Genetic studies suggest that non–cystic
sputum obtained from patients with primary fibrosis bronchiectasis is associated with a spec-
ciliary dyskinesia also is abnormally hypercon- trum of genetic risks, including abnormal muco-
centrated, which suggests that a “second hit” sal host defense, immune function, or connec-
contributes to the pathogenesis of the disease tive tissue.60 The recent reports that sputum
(Fig. 1C).58 Studies that identified the mechano- obtained from patients with non–cystic fibrosis
transduction pathways linking mucus concen- bronchiectasis is hyperconcentrated suggest that
tration with cilial strain–induced ATP release an epithelial defect in the regulation of mucin
also showed that this signaling pathway was hydration may contribute to disease pathogene-
defective in patients with primary ciliary dyski- sis (Fig. 1C). Currently, genetic studies have not
nesia.24 This defect in ciliary sensing probably linked ion-transport genes to non–cystic fibrosis
reduces the release of ATP onto airway surfaces, bronchiectasis, and direct measurements have
thereby decreasing fluid secretion and generat- suggested that CFTR functions normally in pa-
ing a hyperconcentrated mucus plaque or plug tients with non–cystic fibrosis bronchiectasis
component to airflow obstruction. without CFTR mutations.63
The pulmonary phenotype of patients with The microbiome of patients with non–cystic
primary ciliary dyskinesia, aside from the more fibrosis bronchiectasis is typical of muco-obstruc-
basilar distribution of bronchiolectasis and bron- tive diseases, characterized by polymicrobial oral
chiectasis, is similar to that of cystic fibrosis but anaerobic bacteria, staphylococcus, and H. influ-
is shifted toward older ages with respect to dis- enzae, with approximately 15 to 20% of patients
ease severity–age relationships.3,59 Notable simi- positive for P. aeruginosa.45,64 In this context, non–
larities to cystic fibrosis include the presence of cystic fibrosis bronchiectasis resembles early cys-
anaerobes in early primary ciliary dyskinesia, the tic fibrosis.50 Patients with non–cystic fibrosis
subsequent acquisition of classic muco-obstruc- bronchiectasis have an incidence of nontubercu-
tive or bronchiectatic pathogens (e.g., staphylo- lous mycobacteria infection that may be as much
coccus, Haemophilus influenzae, and P. aeruginosa), as 60% higher than the incidence among pa-
and severe neutrophilic inflammation in the air- tients with other muco-obstructive diseases; this
ways with raised DNA concentrations.3 It is not finding indicates a potential role of immune-
clear whether the milder disease phenotype of deficiency alleles in non–cystic fibrosis bronchi-
patients with primary ciliary dyskinesia reflects ectasis.60,65
the retention of partial ciliary activity in many
genotypes of the disease, the persistence of Di agnosis
CFTR functions other than hydration in primary
ciliary dyskinesia, or other factors. Disease-specific criteria assist in the diagnosis
of each muco-obstructive lung disease: for cystic
Non–Cystic Fibrosis Bronchiectasis fibrosis, levels of chloride anions in sweat and
Non–cystic fibrosis bronchiectasis is a phenotype genetic testing66; for COPD, exposure history
defined by bronchiectasis in the absence of a and spirometry1; for primary ciliary dyskinesia,
monogenetic cause.60 Although non–cystic fibrosis nasal nitric-oxide measurements, cilia waveform
bronchiectasis is now typically defined by dilated analyses, and genetic testing3,57; and for non–
bronchi on CT, classic pathological studies have cystic fibrosis bronchiectasis, CT scanning.60
highlighted severe small-airway disease, includ- The tools to make a general diagnosis of muco-
ing bronchiolectasis, mucus plugging, and in- obstructive disease are also available. Patient-
flammation, in this syndrome.61 Consistent with related outcomes regarding the presence of
these pathological findings, non–cystic fibrosis cough, sputum production, and exacerbation fre-
bronchiectasis has the typical small-airway air- quency can be obtained from general respiratory
flow impairment of muco-obstructive diseases.62 questionnaires.5 Measures of sputum inflamma-
Non–cystic fibrosis bronchiectasis probably tory cells and cytokines or chemokines aid in
reflects an interaction between environmental establishing the inflammatory component of
stresses and genetic host-defense risk alleles, airway muco-obstruction.

1950 n engl j med 380;20 nejm.org  May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

Measures of mucus concentration (percent described in cystic fibrosis, producing sustained


solids) and total mucin concentrations have also increases in mucociliary clearance, increased FEV1,
been explored as diagnostic tools.10 For example, and reduced frequencies of exacerbation.69,71
mucin concentrations in sputum are signifi- Inhalation of hypertonic saline is often used in
cantly associated with chronic bronchitis symp- patients with non–cystic fibrosis bronchiectasis
toms defined by patient-related outcome data, or primary ciliary dyskinesia, with some early
and receiver-operating-characteristic curve analy- evidence of efficacy,72 whereas inhalation of hy-
ses suggest that mucin concentration is a good pertonic saline appears to be safe in sputum
biomarker for COPD.10 induction in patients with COPD but is untested
as yet therapeutically.73
Ther a pie s for Muc o - Obs t ruc t i v e
Dise a se s Mucolytics
Two recent observations suggest that attacking
Disease-specific therapies for one muco-obstruc- abnormal mesh and gel properties of mucus in
tive disease, cystic fibrosis, are now available. addition to reducing concentration may be thera-
Ivacaftor (VX-770) is a potentiator of residual peutic. First, mucus viscosity is a key variable
CFTR function that has been approved for pa- governing the mucus cohesive and adhesive prop-
tients with cystic fibrosis with gating and some erties relevant to cough efficiency, and reduc-
splicing CFTR mutations.67 Ivacaftor provided a tions in viscosity with mucin S–S bond–reducing
model for the development of therapies for muco- or surfactant agents decrease mucus adhesion
obstruction, as evidenced by impressive associa- and cohesion independent of concentration.11
tions between improved airway-surface hydration Second, the mucus plaques or flakes that are
in vitro, peripheral and central mucociliary clear- recovered from young patients with cystic fibro-
ance in vivo, and clinically relevant outcomes sis during bronchoalveolar lavage are unable to
(e.g., forced expiratory volume in 1 second [FEV1]). swell and dissolve in excess solvent47 (i.e., they
For patients with cystic fibrosis who are homo- are “permanent” gels). Permanency can be con-
zygous for a deletion of phenylalanine 508 ferred by the generation of bonds with a long
(F508del) in CFTR, ivacaftor–lumacaftor and half-life between mucin polymers that can reflect
tezacaftor–ivacaftor are available but offer more oxidant-induced inter-mucin S–S bonds, inflam-
modest clinical benefit. Three-drug combinations matory protein-mediated “sticker” interactions
of correctors and potentiators await approval but between adjacent mucins, or both.74 Thus, ther-
appear to be highly effective in patients with at apies that are designed to reduce inter-mucin
least one F508del mutation (approximately 90% S–S bonds or block mucin–sticker interactions
of patients). (e.g., with surfactants or glycopolymers) are be-
ing developed to improve both mucus swelling
Hydrators and cough clearance.8,75 Inhaled acetylcysteine has
Perhaps the most direct approach in the treat- not proved to have the mucin-reductive activity
ment of muco-obstructive lung diseases is to required for a therapeutic effect.8 (Additional
reduce the concentration of pathologic mucus references for this article are listed in the Sup-
— that is, rehydrate it. The currently available plementary Appendix.)
approach to achieve this goal is inhalation of
osmotically active aerosols (e.g., hypertonic saline C onclusions
or mannitol).68,69 In clinical development are
modulators of ion transport that may redirect Muco-obstructive diseases are characterized
airway epithelial ion transport from net absorp- by mucus hyperconcentration. The four muco-­
tive to secretory directions, providing a mecha- obstructive diseases differ with respect to the
nism for epithelial restoration of airway-surface epithelial abnormalities that produce mucus hyper­
hydration.70 concentration but follow a final common path
Hypertonic saline is the most widely tested of mucus concentration–dependent formation
inhaled osmotic agent in muco-obstructive lung of mucus plaques and plugs. Adherent mucus
diseases. The use of hypertonic saline is best plaques may stimulate a positive-feedback muco-

n engl j med 380;20 nejm.org  May 16, 2019 1951


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

inflammatory cycle that renders mucus plaques efficiency may be poor. However, the concept that
permanent and damaging to airway walls. Thera- these diseases may be highly treatable emanates
pies that are designed to rehydrate and restore from studies involving patients with cystic fibro-
mucous viscous or elastic properties are rational. sis in whom potentiators have reversed mucus
The challenge is to deliver these therapies to the obstruction in both large and small airways.67
small airways, where mucus obstruction may be Disclosure forms provided by the author are available with the
complete and the physics of aerosol-deposition full text of this article at NEJM.org.

References
1. Han MK, Agusti A, Calverley PM, et al. meric mucins in airways mucus. Annu Rev 28. Cole PJ. Inflammation: a two-edged
Chronic obstructive pulmonary disease Physiol 2008;​70:​459-86. sword — the model of bronchiectasis. Eur
phenotypes: the future of COPD. Am J 15. Rubinstein M, Colby RH. Polymer J Respir Dis Suppl 1986;​147:​6-15.
Respir Crit Care Med 2010;​182:​598-604. physics. Oxford, United Kingdom:​Oxford 29. Tyson JJ, Chen KC, Novak B. Sniffers,
2. King PT, Holdsworth SR, Freezer NJ, University Press, 2003. buzzers, toggles and blinkers: dynamics of
Villanueva E, Holmes PW. Characterisa- 16. Anderson WH, Coakley RD, Button B, regulatory and signaling pathways in the
tion of the onset and presenting clinical et al. The relationship of mucus concen- cell. Curr Opin Cell Biol 2003;​15:​221-31.
features of adult bronchiectasis. Respir tration (hydration) to mucus osmotic pres- 30. Chalmers JD, Moffitt KL, Suarez-
Med 2006;​100:​2183-9. sure and transport in chronic bronchitis. Cuartin G, et al. Neutrophil elastase ac-
3. Knowles MR, Zariwala M, Leigh M. Am J Respir Crit Care Med 2015;​192:​182- tivity is associated with exacerbations and
Primary ciliary dyskinesia. Clin Chest Med 90. lung function decline in bronchiectasis.
2016;​37:​449-61. 17. Lucas A, Douglas LC. Principles under- Am J Respir Crit Care Med 2017;​195:​1384-
4. Rowe SM, Miller S, Sorscher EJ. Cystic lying ciliary activity in the respiratory 93.
fibrosis. N Engl J Med 2005;​ 352:​ 1992- tract. Arch Otolaryngol 1934;​20:​518-41. 31. Sly PD, Gangell CL, Chen L, et al. Risk
2001. 18. Okuda K, Chen G, Subramani DB, factors for bronchiectasis in children with
5. Jones PW, Forde Y. St George’s respi- et al. Localization of secretory mucins cystic fibrosis. N Engl J Med 2013;​368:​
ratory questionnaire manual. London:​ MUC5AC and MUC5B in normal/healthy 1963-70.
St George’s, University of London, 2009 human airways. Am J Respir Crit Care 32. Worlitzsch D, Tarran R, Ulrich M,
(http://www​.healthstatus​.sgul​.ac​.uk/​SGRQ Med 2019;​199:​715-27. et al. Effects of reduced mucus oxygen
_download/​SGRQ%20Manual%20June% 19. Zhu Y, Ehre C, Abdullah LH, et al. concentration in airway Pseudomonas in-
202009​.pdf). Munc13-2-/- baseline secretion defect re- fections of cystic fibrosis patients. J Clin
6. Rogers DF. Physiology of airway mu- veals source of oligomeric mucins in mouse Invest 2002;​109:​317-25.
cus secretion and pathophysiology of hy- airways. J Physiol 2008;​586:​1977-92. 33. Cowley ES, Kopf SH, LaRiviere A,
persecretion. Respir Care 2007;​52:​1134- 20. Widdicombe JH, Wine JJ. Airway gland Ziebis W, Newman DK. Pediatric cystic
46. structure and function. Physiol Rev 2015;​ fibrosis sputum can be chemically dy-
7. Dunican EM, Elicker BM, Gierada DS, 95:​1241-319. namic, anoxic, and extremely reduced due
et al. Mucus plugs in patients with asth- 21. Boucher RC. Human airway ion trans- to hydrogen sulfide formation. MBio 2015;​
ma linked to eosinophilia and airflow ob- port: part one. Am J Respir Crit Care Med 6(4):​e00767.
struction. J Clin Invest 2018;​128:​997-1009. 1994;​150:​271-81. 34. Tunney MM, Field TR, Moriarty TF,
8. Fahy JV, Dickey BF. Airway mucus 22. Tarran R, Button B, Picher M, et al. et al. Detection of anaerobic bacteria in
function and dysfunction. N Engl J Med Normal and cystic fibrosis airway surface high numbers in sputum from patients
2010;​363:​2233-47. liquid homeostasis: the effects of phasic with cystic fibrosis. Am J Respir Crit Care
9. Henderson AG, Ehre C, Button B, et al. shear stress and viral infections. J Biol Med 2008;​177:​995-1001.
Cystic fibrosis airway secretions exhibit Chem 2005;​280:​35751-9. 35. Sze MA, Hogg JC, Sin DD. Bacterial
mucin hyperconcentration and increased 23. Leith DE. Cough. Phys Ther 1968;​48:​ microbiome of lungs in COPD. Int J Chron
osmotic pressure. J Clin Invest 2014;​124:​ 439-47. Obstruct Pulmon Dis 2014;​9:​229-38.
3047-60. 24. Button B, Okada SF, Frederick CB, 36. Segal LN, Clemente JC, Tsay JC, et al.
10. Kesimer M, Ford AA, Ceppe A, et al. Thelin WR, Boucher RC. Mechanosensi- Enrichment of the lung microbiome with
Airway mucin concentration as a marker tive ATP release maintains proper mucus oral taxa is associated with lung inflam-
of chronic bronchitis. N Engl J Med 2017;​ hydration of airways. Sci Signal 2013;​6:​ mation of a Th17 phenotype. Nat Micro-
377:​911-22. ra46. biol 2016;​1:​16031.
11. Button B, Goodell HP, Atieh E, et al. 25. Cole AM, Liao HI, Stuchlik O, Tilan J, 37. Flynn JM, Niccum D, Dunitz JM,
Roles of mucus adhesion and cohesion in Pohl J, Ganz T. Cationic polypeptides are Hunter RC. Evidence and role for bacterial
cough clearance. Proc Natl Acad Sci U S A required for antibacterial activity of hu- mucin degradation in cystic fibrosis air-
2018;​115:​12501-6. man airway fluid. J Immunol 2002;​169:​ way disease. PLoS Pathog 2016;​ 12(8):​
12. Rose MC, Voynow JA. Respiratory tract 6985-91. e1005846.
mucin genes and mucin glycoproteins in 26. Cole AM, Dewan P, Ganz T. Innate 38. Matsui H, Wagner VE, Hill DB, et al.
health and disease. Physiol Rev 2006;​86:​ antimicrobial activity of nasal secretions. A physical linkage between cystic fibrosis
245-78. Infect Immun 1999;​67:​3267-75. airway surface dehydration and Pseudo-
13. Button B, Cai LH, Ehre C, et al. A peri- 27. Livraghi-Butrico A, Grubb BR, Wilkin- monas aeruginosa biofilms. Proc Natl
ciliary brush promotes the lung health by son KJ, et al. Contribution of mucus con- Acad Sci U S A 2006;​103:​18131-6.
separating the mucus layer from airway centration and secreted mucins Muc5ac 39. Rosenfeld M, Emerson J, Williams-
epithelia. Science 2012;​337:​937-41. and Muc5b to the pathogenesis of muco- Warren J, et al. Defining a pulmonary
14. Thornton DJ, Rousseau K, McGuckin obstructive lung disease. Mucosal Immu- exacerbation in cystic fibrosis. J Pediatr
MA. Structure and function of the poly- nol 2017;​10:​395-407. 2001;​139:​359-65.

1952 n engl j med 380;20 nejm.org  May 16, 2019

The New England Journal of Medicine


Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Muco-Obstructive Lung Diseases

40. Sethi S, Maloney J, Grove L, Wrona C, pulmonary disease. Physiol Rev 2017;​97:​ et al. Effect of long-term, low-dose eryth-
Berenson CS. Airway inflammation and 529-52. romycin on pulmonary exacerbations
bronchial bacterial colonization in chron- 52. Clunes LA, Davies CM, Coakley RD, among patients with non-cystic fibrosis
ic obstructive pulmonary disease. Am J et al. Cigarette smoke exposure induces bronchiectasis: the BLESS randomized
Respir Crit Care Med 2006;​173:​991-8. CFTR internalization and insolubility, controlled trial. JAMA 2013;​309:​1260-7.
41. Goss CH, Burns JL. Exacerbations in leading to airway surface liquid dehydra- 65. Aksamit TR, O’Donnell AE, Barker A,
cystic fibrosis. 1: Epidemiology and patho- tion. FASEB J 2012;​26:​533-45. et al. Adult patients with bronchiectasis:
genesis. Thorax 2007;​62:​360-7. 53. Cantin AM, Bilodeau G, Ouellet C, a first look at the US Bronchiectasis Re-
42. Boucher RC. On the pathogenesis of Liao J, Hanrahan JW. Oxidant stress sup- search Registry. Chest 2017;​151:​982-92.
acute exacerbations of mucoobstructive presses CFTR expression. Am J Physiol 66. Farrell PM, White TB, Ren CL, et al.
lung diseases. Ann Am Thorac Soc 2015;​ Cell Physiol 2006;​290:​C262-C270. Diagnosis of cystic fibrosis: consensus
12:​Suppl 2:​S160-S163. 54. Kreda SM, Seminario-Vidal L, van guidelines from the Cystic Fibrosis Foun-
43. Lee AL, Button BM, Denehy L, et al. Heusden CA, et al. Receptor-promoted dation. J Pediatr 2017;​181:​Suppl:​S4-S15.e1.
Proximal and distal gastro-oesophageal exocytosis of airway epithelial mucin gran- 67. Ramsey BW, Davies J, McElvaney NG,
reflux in chronic obstructive pulmonary ules containing a spectrum of adenine et al. A CFTR potentiator in patients with
disease and bronchiectasis. Respirology nucleotides. J Physiol 2010;​588:​2255-67. cystic fibrosis and the G551D mutation.
2014;​19:​211-7. 55. Martinez-Garcia MA, Miravitlles M. N Engl J Med 2011;​365:​1663-72.
44. Brodlie M, Aseeri A, Lordan JL, et al. Bronchiectasis in COPD patients: more 68. Anderson SD, Daviskas E, Brannan
Bile acid aspiration in people with cystic than a comorbidity? Int J Chron Obstruct JD, Chan HK. Repurposing excipients as
fibrosis before and after lung transplan- Pulmon Dis 2017;​12:​1401-11. active inhalation agents: the mannitol
tation. Eur Respir J 2015;​46:​1820-3. 56. Murphy TF, Brauer AL, Eschberger K, story. Adv Drug Deliv Rev 2018;​133:​45-
45. Cox MJ, Turek EM, Hennessy C, et al. et al. Pseudomonas aeruginosa in chronic 56.
Longitudinal assessment of sputum micro- obstructive pulmonary disease. Am J 69. Donaldson SH, Bennett WD, Zeman
biome by sequencing of the 16S rRNA Respir Crit Care Med 2008;​177:​853-60. KL, Knowles MR, Tarran R, Boucher RC.
gene in non-cystic fibrosis bronchiectasis 57. Horani A, Ferkol TW, Dutcher SK, Mucus clearance and lung function in cys-
patients. PLoS One 2017;​12(2):​e0170622. Brody SL. Genetics and biology of pri- tic fibrosis with hypertonic saline. N Engl
46. Leung HM, Birket S, Hyun C, et al. mary ciliary dyskinesia. Paediatr Respir J Med 2006;​354:​241-50.
The study of functional microanatomy of Rev 2016;​18:​18-24. 70. Shei RJ, Peabody JE, Kaza N, Rowe SM.
CF airways using clinical intranasal micro- 58. Bush A, Payne D, Pike S, Jenkins G, The epithelial sodium channel (ENaC) as
OCT imaging. Pediatr Pulmonol 2018;​53:​ Henke MO, Rubin BK. Mucus properties a therapeutic target for cystic fibrosis.
Suppl 2:​174. abstract. in children with primary ciliary dyskine- Curr Opin Pharmacol 2018;​43:​152-65.
47. Esther CR, Muhlebach MS, Ehre C, et al. sia: comparison with cystic fibrosis. Chest 71. Elkins MR, Robinson M, Rose BR, et al.
Mucus accumulation in the lungs precedes 2006;​129:​118-23. A controlled trial of long-term inhaled
structural changes and infection in chil- 59. Maglione M, Bush A, Nielsen KG, et al. hypertonic saline in patients with cystic
dren with cystic fibrosis. Sci Transl Med Multicenter analysis of body mass index, fibrosis. N Engl J Med 2006;​354:​229-40.
2019;​11(486):​eaav3488. lung function, and sputum microbiology 72. Paff T, Daniels JM, Weersink EJ, Lutter
48. Simel DL, Mastin JP, Pratt PC, et al. in primary ciliary dyskinesia. Pediatr Pul- R, Vonk Noordegraaf A, Haarman EG. A
Scanning electron microscopic study of monol 2014;​49:​1243-50. randomised controlled trial on the effect
the airways in normal children and in 60. Flume PA, Chalmers JD, Olivier KN. of inhaled hypertonic saline on quality
patients with cystic fibrosis and other Advances in bronchiectasis: endotyping, of life in primary ciliary dyskinesia. Eur
lung diseases. Pediatr Pathol 1984;​2:​47- genetics, microbiome, and disease hetero- Respir J 2017;​49:​49.
64. geneity. Lancet 2018;​392:​880-90. 73. Fujimoto K, Yasuo M, Urushibata K,
49. Abdullah LH, Coakley R, Webster MJ, 61. Whitwell F. A study of the pathology Hanaoka M, Koizumi T, Kubo K. Airway
et al. Mucin production and hydration re- and pathogenesis of bronchiectasis. Tho- inflammation during stable and acutely
sponses to mucopurulent materials in nor- rax 1952;​7:​213-39. exacerbated chronic obstructive pulmo-
mal versus cystic fibrosis airway epithe- 62. Roberts HR, Wells AU, Milne DG, et al. nary disease. Eur Respir J 2005;​25:​640-6.
lia. Am J Respir Crit Care Med 2018;​197:​ Airflow obstruction in bronchiectasis: cor- 74. Yuan S, Hollinger M, Lachowicz-
481-91. relation between computed tomography Scroggins ME, et al. Oxidation increases
50. Muhlebach MS, Zorn BT, Esther CR, features and pulmonary function tests. mucin polymer cross-links to stiffen air-
et al. Initial acquisition and succession of Thorax 2000;​55:​198-204. way mucus gels. Sci Transl Med 2015;​7:​
the cystic fibrosis lung microbiome is as- 63. Bienvenu T, Sermet-Gaudelus I, Burgel 276ra27.
sociated with disease progression in infants PR, et al. Cystic fibrosis transmembrane 75. Tarrant BJ, Le Maitre C, Romero L, et al.
and preschool children. PLoS Pathog 2018;​ conductance regulator channel dysfunc- Mucoactive agents for chronic, non-cystic
14(1):​e1006798. tion in non-cystic fibrosis bronchiectasis. fibrosis lung disease: a systematic review
51. Hogg JC, Paré PD, Hackett TL. The Am J Respir Crit Care Med 2010;​181:​1078- and meta-analysis. Respirology 2017;​22:​
contribution of small airway obstruction 84. 1084-92.
to the pathogenesis of chronic obstructive 64. Serisier DJ, Martin ML, McGuckin MA, Copyright © 2019 Massachusetts Medical Society.

n engl j med 380;20 nejm.org  May 16, 2019 1953


The New England Journal of Medicine
Downloaded from nejm.org by ADRIANA FONSECA on June 13, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.

You might also like