You are on page 1of 15

[ Recent Advances in Chest Medicine ]

Current Concepts in Pathogenesis,


Diagnosis, and Management of
Smoking-Related Interstitial Lung Diseases
Anupam Kumar, MD; Sujith V. Cherian, MD; Robert Vassallo, MD; Eunhee S. Yi, MD; and Jay H. Ryu, MD

Tobacco exposure results in various changes to the airways and lung parenchyma. Although
emphysema represents the more common injury pattern, in some individuals, cigarette smoke
injures alveolar epithelial cells and other lung cells, resulting in diffuse infiltrates and paren-
chymal fibrosis. Smoking can trigger interstitial injury patterns mediated via recruitment and
inappropriate persistence of myeloid and other immune cells, including eosinophils. As our
understanding of the role of cigarette smoke constituents in triggering lung injury continues to
evolve, so does our recognition of the spectrum of smoking-related interstitial lung changes.
Although respiratory bronchiolitis-interstitial lung disease, desquamative interstitial pneu-
monia, pulmonary Langerhans cell histiocytosis, and acute eosinophilic pneumonia have a well-
established association with tobacco use, its role and impact on idiopathic pulmonary fibrosis,
combined pulmonary fibrosis and emphysema, and connective tissue disease-related intersti-
tial lung diseases is still ambiguous. Smoking-related interstitial fibrosis is a relatively newly
appreciated entity with distinct histopathologic features but with unclear clinical ramifications.
Increased implementation of lung cancer screening programs and utilization of CT scans in
thoracic imaging have also resulted in increased identification of “incidental” or “subclinical”
interstitial lung changes in smokers, the ensuing impact of which remains to be studied.
CHEST 2018; 154(2):394-408

KEY WORDS: interstitial lung disease; pulmonary fibrosis; smoking

Cigarette smoking is implicated to varying diseases in this spectrum can be grouped into
degrees in a distinct but heterogeneous those that likely have a causal association
group of parenchymal lung diseases with tobacco exposure (respiratory
(smoking-related interstitial lung diseases bronchiolitis-interstitial lung disease
[ILDs]) in addition to emphysema. Lung [RB-ILD], desquamative interstitial

ABBREVIATIONS: AEC2 = type 2 alveolar epithelial cells; AEP = acute Pulmonary, Critical Care and Sleep Medicine (Dr Cherian), University
eosinophilic pneumonia; CPFE = combined pulmonary fibrosis and of Texas Health Science Center at Houston-McGovern Medical School,
emphysema; CTD-ILD = connective tissue disease-related interstitial Houston, TX; and the Division of Pulmonary and Critical Care
lung disease; DIP = desquamative interstitial pneumonia; GGO = Medicine (Drs Vassallo and Ryu) and Department of Laboratory
ground-glass opacity; HRCT = high-resolution CT; ILD = interstitial Medicine and Pathology (Dr Yi), Mayo Clinic College of Medicine and
lung disease; IPF = idiopathic pulmonary fibrosis; PDGF = platelet- Science, Rochester, MN.
derived growth factor; PH = pulmonary hypertension; PLCH = pul- CORRESPONDENCE TO: Anupam Kumar, MD, Division of Pulmonary
monary Langerhans cell histiocytosis; RA = rheumatoid arthritis; RB = and Critical Care Medicine, Spectrum Health-Michigan State
respiratory bronchiolitis; RB-ILD = respiratory bronchiolitis-interstitial University College of Human Medicine, 100 Michigan St NE, Grand
lung disease; SRIF = smoking-related interstitial fibrosis; UIP = usual Rapids, MI 49503; e-mail: anupamkumarmd@gmail.com
interstitial pneumonia Copyright Ó 2017 American College of Chest Physicians. Published by
AFFILIATIONS: From the Division of Pulmonary and Critical Care Elsevier Inc. All rights reserved.
Medicine (Dr Kumar), Spectrum Health-Michigan State University DOI: https://doi.org/10.1016/j.chest.2017.11.023
College of Human Medicine, Grand Rapids, MI; Division of

394 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


pneumonia [DIP], pulmonary Langerhans cell enzyme system that performs the essential function of
histiocytosis [PLCH], and acute eosinophilic pneumonia maintaining telomeres on the ends of chromosomes.3
[AEP]) and those in which smoking likely acts in This cellular function is critical as evidenced by the
conjunction with endogenous or exogenous factors but predisposition of cells with abnormally short telomeres
causality is yet to be established (idiopathic pulmonary to develop a phenotype of senescence and apoptosis.
fibrosis [IPF], combined pulmonary fibrosis and Studies performed in telomerase knockout mice revealed
emphysema [CPFE], and connective tissue disease- an increased susceptibility to cigarette smoke-induced
related interstitial lung disease [CTD-ILD]). The present damage and development of emphysema.4 This finding
review discusses our current understanding of the may be particularly relevant to understanding
manifold mechanisms by which cigarette smoke susceptibility to both emphysema and fibrosis because of
inhalation can trigger and propagate interstitial lung the demonstration that up to 10% of patients with
injury in smokers, as well as the key clinical features that nonfamilial fibrotic idiopathic interstitial pneumonia
differentiate these disorders. We also discuss the (most of whom have IPF have abnormally short
potential clinical ramifications of smoking-related telomere lengths5). Although this study did not perform
interstitial fibrosis (SRIF) and the downstream impact of correlations between smoking history and telomere
incidentally detected “subclinical” interstitial lung length, it does suggest a common mechanism by which
changes in smokers. certain smokers may develop a predisposition to alveolar
epithelial cell senescence and an abnormal lung
Mechanisms and Spectrum of Cigarette regenerative pathway that may lead to the phenotype of
Smoke-Induced ILDs emphysema in some and fibrosis in others (or even both
in some patients).
Tobacco smoke is a toxic assortment of particulate
matter and various chemicals that induce inflammatory, The induction of alveolar epithelial cellular senescence
genotoxic, proliferative, and other stimuli to lung cells by cigarette smoke is potentially also pivotal for the
that extend from the conducting to the gas exchange development of either emphysema or interstitial changes
zones. Reactive oxidant species, nicotine, and other and fibrosis in certain smokers. Cellular senescence, a
toxins in cigarette smoke induce epithelial as well as state of replicative arrest brought on by cellular stressors
endothelial cellular activation, often leading to secretion (including cigarette smoke), results in emergence of the
of a variety of inflammatory cytokines and chemokines senescence-associated secretory phenotype.6 The
that secondarily recruit immune cells into various lung mediators secreted by these cells include a variety of
compartments, including small and large airways and cytokines, chemokines, matrix metalloproteinases, and
the lung parenchyma.1,2 Emphysema, the more common growth factors that have been implicated in the
parenchymal lung injury pattern associated with pathogenesis of both emphysema and pulmonary
smoking, is associated with alveolar cell apoptosis and fibrosis.6 It is possible that the induction of senescent
destruction accompanied by loss of lung scaffolding. cells in the lungs of predisposed smokers may be
Although morphologically very different compared with sufficient to drive a subsequent inflammatory and tissue
lesions observed in smoking-induced diffuse lung remodeling response, such that the ensuing
diseases, some of the mechanisms by which smoking inflammatory cell recruitment becomes a secondary
induces emphysema are similar to those involved in phenomenon of the aberrant biology, rather than a
fibrosis and distal lung remodeling. primary driver of tissue injury, and may provide some
insight into why antiinflammatory (corticosteroid or
One cell type that is likely central in the pathogenesis of
other immunomodulatory) therapy is generally lacking
both emphysema and lung fibrosis is the alveolar
in therapeutic efficacy in at least some of the tobacco-
epithelial stem cell. In the lung, a subset of type 2
induced lung diseases.
alveolar epithelial cells (AEC2) performs the function of
new AEC2 progenitors as well as a source of new type 1 Cigarette smoke, or its constituents, can also directly
alveolar epithelial cells. In this context, certain induce profibrotic factors from lung cells.7,8 Among the
pathogenic events such as telomere dysfunction and growth factors directly induced by cigarette smoke,
predisposition to cellular senescence may provide transforming growth factor-beta 1 (TGF-b1) and
insight into the susceptibility of some smokers to platelet-derived growth factor (PDGF)-A and -B are
develop either emphysema or fibrotic interstitial lung particularly relevant.8,9 TGF-b1 is essential for matrix
changes (or a combination of both). Telomerase is an generation (including collagen, fibronectin, and elastin)

chestjournal.org 395
by fibroblasts and myofibroblasts. PDGF induces cells. Studies on BAL samples of patients with AEP have
fibroblast and myofibroblast proliferation and thus may also shown increased levels of IL-5 and IL-33, both
amplify TGF-b1 effects by enhancing the number of cells potent stimulators of eosinophil recruitment, activation,
available to respond and produce matrix proteins. and proliferation.16,17 IL-5 levels were also found to
Immunohistochemical studies performed on lung biopsy inversely correlate with peripheral eosinophilia,
specimens from patients with smoking-induced diffuse suggesting its role in recruiting eosinophils from
lung diseases have shown upregulation of both TGF-b1 peripheral blood into the lungs, and that an increase in
as well as PDGF, suggesting a role for these factors in blood eosinophil count in a patient with AEP is likely a
tissue remodeling and fibrosis.10,11 marker of resolving inflammation.16,18 The eosinophil
accumulation in AEP causes widespread capillary
Numerous lines of evidence now also support a key role
endothelial damage and increased vascular permeability,
for osteopontin, a glycoprotein with cytokine-like
and it results in diffuse alveolar damage with interstitial
properties, in the pathogenesis of smoking-induced
eosinophil infiltration. Although cigarette smoke is
diffuse lung diseases.12,13 Immunohistochemical studies
certainly a stimulus for this eosinophil-mediated lung
on PLCH and DIP lung biopsy specimens revealed
injury, other extrinsic stimuli can also plausibly
strong osteopontin immunoreactivity in infiltrating
stimulate such a response pattern.19
macrophages and CD1aþ cells, as well as AEC2 cells.12
Functionally, osteopontin may be critically involved in One of the most compelling arguments linking the
antigen-presenting cell trafficking into the lung and may diseases listed in Table 1 with cigarette smoking is the
also play a critical role in the development of T-helper- co-existence of the various aforementioned patterns of
17 polarized responses that subsequently drive interstitial and airway injuries in the same patient.20
inflammation in certain tobacco-induced lung Although smoking clearly can cause these diverse lung
diseases.13 Osteopontin, along with other cytokines (eg, injury patterns, it is not certain why some smokers
granulocyte-macrophage colony stimulating factor, develop one specific entity rather than another lesion.
TGF-b1) can stimulate the recruitment of Langerhans These diverse lung responses to cigarette smoke imply
cells.10,14 Exposure to tobacco smoke may also enhance that there must be additional co-factors (either
the survival of myeloid cells, including dendritic cells, by endogenous/genetic or exogenous) that result in the
inducing cellular response proteins involved in the induction of a specific disease phenotype in a given
regulation of cell death by apoptosis.15 Recruitment of smoker. Similarly, the extent to which any of these
myeloid cells with morphologic features similar to smoking-induced diffuse lung diseases progress to lung
Langerhans cells around small airways is likely an early remodeling and airway and/or parenchymal fibrosis
event that precedes secondary recruitment of other varies considerably between different individuals and
inflammatory cells and results in formation of the cannot simply be explained by cumulative tobacco
bronchiolocentric granulomatous-like lesions exposure, implying the need to identify further genetic
characteristic of PLCH. factors that interplay with smoking in the determination
of the eventual disease phenotype.
Although the potential mechanisms mentioned earlier
provide some insights into the pathogenesis of chronic ILDs Causally Associated With Smoking
forms of lung injury such as fibrosis and emphysema, it ILDs in which cigarette smoking has been causally
has also become apparent that cigarette smoke can implicated include RB-ILD, DIP, PLCH, and AEP. SRIF
induce an acute form of lung injury through eosinophil and “subclinical” interstitial changes in smokers are also
recruitment and activation in some patients, resulting in included in this category. The clinical features, including
AEP. Constituents of the cytoplasmic granules of the demographic characteristics and pattern of pulmonary
eosinophil such as eosinophilic cationic proteins, function test results in these disorders, are summarized
leukotrienes, and other cytokines cause local tissue in Table 1.
injury and edema of lungs through a variety of
mechanisms. Exaggerated eosinophil response and the Respiratory Bronchiolitis-ILD
release of these substances can also stimulate mast cell Respiratory bronchiolitis (RB) is a histopathologic term
differentiation and T lymphocyte proliferation.16 that refers to the deposition of pigmented macrophages
Prospective studies on patients with AEP also found in the respiratory bronchioles and peribronchiolar
increased tryptase levels, confirming the role of mast alveolar spaces.21 Although RB can be considered a

396 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


chestjournal.org

TABLE 1 ] Summary of Significant Clinical Characteristics of Smoking-Related ILDs


Demographic
Smoking-Related ILD Characteristics Symptoms PFT Results Radiology (CT Scan Features) Key Management Strategies
RB-ILD M ¼ F, 30-60 y Cough, dyspnea Normal, obstructive, GGO  centrilobular nodules Smoking cessation,
restrictive, or mixed (usually upper lobe) corticosteroids in
refractory/progressive
cases
DIP M > F, 40-60 y Cough, dyspnea, hemoptysis Normal, obstructive, GGO with lower lobe Smoking cessation,
(rare) restrictive, or mixed. predominance corticosteroids in
Reduced DLCO (centrilobular nodules are refractory/progressive
rare) cases
SRIF M ¼ F, 40-60 y Cough, dyspnea Obstructive, restrictive, or Centrilobular nodules  GGO Smoking cessation, consider
(diagnosed only with mixed (similar to RB-ILD) steroids in severe cases
histopathology) (no data)
PLCH M ¼ F, 20-40 y Cough, dyspnea, pneumothorax Restriction (initial) and Upper lobe nodules, mix of Smoking cessation.
extrapulmonary (diabetes obstruction (advanced) nodules and bizarre- Cladribine in severe
insipidus, bone cysts) shaped cysts (advanced disease. Consider lung
stages), sparing of transplantation in
costophrenic angle advanced cases
AEP M > F, 20-40 y Acute onset of cough, dyspnea, May not be obtained in acute Bilateral, often diffuse GGO Smoking cessation and
and fever. Respiratory failure illness but can be and reticular opacities. corticosteroids (excellent
in severe cases obstructive, restrictive, or Pleural effusion may be response)
mixed seen
IPF M > F, > 60 y Progressive dyspnea, dry cough Restrictive Basal reticulation, traction Smoking cessation, consider
bronchiectasis, antifibrotic therapy (if UIP/
honeycombing IPF confirmed) or lung
transplantation in suitable
candidates
CPFE M > F, > 60 y Progressive dyspnea, cough Pseudo-normal PFT with Upper lobe emphysema with Smoking cessation, consider
disproportionately low lower lobe fibrosis lung transplantation in
DLCO suitable candidates

AEP ¼ acute eosinophilic pneumonia; CPFE ¼ combined pulmonary fibrosis and emphysema; DIP ¼ desquamative interstitial pneumonia; DLCO ¼ diffusing capacity of the lung for carbon monoxide; F ¼ female;
GGO ¼ ground-glass opacity; ILD ¼ interstitial lung disease; IPF ¼ idiopathic pulmonary fibrosis; M ¼ male; PFT ¼ pulmonary function test; PLCH ¼ pulmonary Langerhans cell histiocytosis; RB-ILD ¼ respiratory
bronchiolitis-interstitial lung disease; SRIF ¼ smoking-related interstitial fibrosis; UIP ¼ usual interstitial pneumonia.
397
tell-tale sign of exposure to tobacco, not all patients with nonspecific changes in other cell lines. Lymphocytic
RB develop clinical manifestations. Thus, RB-ILD is predominance on BAL in a patient with centrilobular
distinct from RB in that it is a form of interstitial nodules and subtle GGOs should prompt consideration
pneumonia with characteristic clinical, radiologic, and of hypersensitivity pneumonitis over RB-ILD.
pathologic features (Table 1).22 Almost all patients with Transbronchial biopsy is not a reliable diagnostic
RB-ILD are current or previous smokers.23-25 However, modality due to the patchy nature of lung involvement.
the intensity of tobacco exposure can be variable with
Histopathology of RB-ILD shows the presence of
occurrence of symptoms even in patients with minimal
brown-pigmented macrophages in the distal airways (ie,
exposure.23 It is also interesting to note that RB is a
respiratory bronchioles and adjacent alveolar spaces)
frequent pathologic finding in smokers (in up to 90% of
(Fig 1B). The distribution is patchy and without overt
patients) who develop spontaneous or recurrent
evidence of fibrosis, although mild peribronchiolar
pneumothorax.26 Aside from cigarette exposure, other
fibrosis may be observed.22,31,32 In addition, patchy
fumes such as solder and fiberglass have also been
lymphocytic and histiocytic infiltration of the
implicated in RB-ILD, albeit rarely.24,27
peribronchiolar interstitium may be seen.31 Whether RB
RB-ILD is significantly better characterized on high- can be distinguished from RB-ILD based on pathologic
resolution CT (HRCT) scans than plain radiographs. findings is controversial, but RB-ILD tends to have more
Characteristic HRCT abnormalities include patchy alveolar fibrosis than RB.33 Thus, in a patient with
ground-glass opacities (GGOs), with or without the compatible clinicoradiologic features and RB-like
presence of fine centrilobular nodules (Fig 1A). In a histopathologic changes, a diagnosis of RB-ILD can be
single center-based study, bronchial thickening (central safely secured.
and peripheral) was the most common finding (90%),
Smoking cessation is the most important
followed by centrilobular nodules (71%) and ground-
recommendation for a patient with RB-ILD. However,
glass opacification (67%).25 Other changes that may be
clinical and physiological improvement following
seen include areas of air trapping and emphysema.23,25 It
smoking cessation is delayed and often
is unusual to see honeycombing or other changes that
incomplete.23,24,34 In a long-term follow-up study by
signify fibrosis such as traction bronchiolectasis/
Portnoy et al,34 75% of the patients survived > 7 years
bronchiectasis. The lung parenchymal changes in RB-
but with clinical and physiological improvement in only
ILD do not have a zonal predominance. This finding is
28% and 10.5% of patients, respectively. In patients with
in contrast to DIP, which also presents with diffuse
worsening lung disease, or lack of improvement,
ground-glass opacification but with lower zone
particularly if the clinical presentation (ie, demographic
predominance, although not universal.28-30
characteristics, radiology findings) is atypical, a surgical
A history of smoking with the aforementioned lung biopsy should be considered for diagnostic
radiographic changes is usually sufficient to diagnose clarification. Second-line treatment for persistent or
RB-ILD. Typical BAL studies reflect an increased progressive RB-ILD is less well established but should be
number of macrophages (as in smokers in general) with attempted in the absence of improvement.

Figure 1 – A, High-resolution CT (HRCT) scan of respiratory bronchiolitis-interstitial lung disease. HRCT scan of the chest of a 42-year-old male
smoker demonstrates patchy ground-glass opacities bilaterally, more prominent in the left lung anteriorly. B, Histopathology of respiratory
bronchiolitis-interstitial lung disease. Mild increase in pigmented alveolar macrophages within the bronchiolar lumen and adjacent alveolar spaces with
minimal interstitial fibrosis. Hematoxylin and eosin staining, 100 original magnification.

398 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


Corticosteroids are often prescribed for individuals with impairment tends to be more severe than in RB-ILD
progressive RB-ILD, although the evidence that supports with at least a moderate impairment in gas transfer (as
such use is sparse.23,34 A dose in the range of 0.5 to gauged by a reduced diffusing capacity of the lung for
1 mg/kg of prednisone is typically started with a gradual carbon monoxide).23,31,35 HRCT scanning shows
taper over months based on benefits and tolerance. extensive bilateral GGOs, which may have a peripheral
Risks/benefits of long-term therapy with corticosteroids and basal predominance (Fig 2A). Unlike in RB-ILD,
do not have supportive data and should be carefully centrilobular nodules are rarely seen.29 Linear opacities
individualized for patients. Some patients may exhibit and reticulation may be seen in up to 60% of patients.30
worsening of clinical and physiological parameters Long-term radiographic follow-up of DIP may
despite treatment with corticosteroids and other demonstrate radiographic evolution with development
immunosuppressant agents.23,24,34 In patients with co- of honeycombing in up to 10% to 20% of patients.29,44
existing features of air trapping, obstructive lung disease, Thus, the distribution of interstitial abnormalities in DIP
or emphysema, inhaled corticosteroids and may sometimes be similar to that seen in usual
bronchodilators could also be considered. interstitial pneumonia (UIP), but the degree of ground-
glass opacification and the relative paucity of
Desquamative Interstitial Pneumonia honeycomb changes in DIP enables distinction from
UIP.29 Bronchoscopy, as in cases of RB-ILD, is used
DIP was first described by Liebow et al35 in 1965, the
primarily to rule out an alternative diagnosis. Kawabata
name originally referring to “desquamation” of epithelial
et al45 reported marked eosinophilia (mean: 18%) and
cells. However, the abnormal collections of cells
moderate neutrophilia (mean: 11%) in the BAL fluid of
observed within the alveolar spaces in this disorder are
85% of the patients. Attempts at obtaining tissue
now known to be accumulations of macrophages and
diagnosis (surgical biopsy is preferred over
not desquamated epithelial cells. Hence the term “DIP”
transbronchial biopsy) should be reserved for ambiguity
is considered by many to be a misnomer.
in diagnosis or disease worsening despite conventional
DIP, like RB-ILD, has a strong association with current treatment.
and previous smoking.30,36 Based on data from several
Pathology of DIP overlaps with that of RB-ILD, the most
studies, 60% to 90% of DIP cases are reported in
striking feature being accumulation of pigmented
smokers.30,32,36 However, in contrast to RB-ILD, which
macrophages in the small airways and alveoli (Fig 2B).
is almost exclusively a smoker’s disease, DIP has been
Key features that differentiate DIP and RB-ILD include
reported secondary to other causes such as connective
the generally uniform and widespread involvement in
tissue diseases (eg, rheumatoid arthritis [RA], systemic
DIP, whereas the macrophage accumulation and fibrotic
sclerosis),37,38 occupational exposure (eg, mycotoxins,
changes center along the small airways and adjacent
dust, solder fumes, flock-workers),24,39-41 drugs (eg,
alveoli (bronchiolocentric) in RB-ILD.31,32 Alveolar
sirolimus),42 and infections.43
septal thickening occurs due to interstitial inflammation.
Symptoms of patients with DIP are similar to RB-ILD Mild to moderate fibrosis may be present. In contrast to
(Table 1). However, the pulmonary physiological UIP, however, patchiness of fibrosis or temporal

Figure 2 – A, HRCT image of desquamative interstitial pneumonia. HRCT scan of the chest of a 32-year-old female smoker demonstrates extensive
patchy ground-glass opacities bilaterally in both lower lobes. B, Histopathology of desquamative interstitial pneumonia. Diffuse and prominent
accumulation of pigmented alveolar macrophages with mild to moderate interstitial fibrosis. Variable amount of chronic inflammatory infiltrates and
lymphoid hyperplasia may be seen (not shown). Hematoxylin and eosin staining, 100 original magnification. See Figure 1 legend for expansion of
abbreviation.

chestjournal.org 399
heterogeneity with fibroblast foci is not a feature of DIP, develops.49,50 BRAF-V600E and other activating cell
allowing relatively easy differentiation. cycle mutations may drive disease activity even in
subjects who quit smoking, which may explain the
In general, most patients with DIP have a good
observation of disease progression in certain individuals
prognosis, especially if smoking cessation is attained
despite smoking cessation.
early in the disease course. Within the limitations of lack
of robust data, results from existing studies suggest an Up to 10% of patients with PLCH can be asymptomatic,
overall survival approaching 90% at 10 years.23,36 and 15% may present with recurrent pneumothorax.51,52
However, progression to respiratory failure is more Aside from pulmonary parenchymal involvement,
likely for patients with DIP than with RB-ILD.23 pulmonary vascular involvement resulting in pulmonary
Whether DIP can progress to UIP is controversial, but hypertension (PH) seems to be relatively common in
follow-up imaging over the long term may show patients with advanced PLCH.53 For instance, in a
evidence of honeycombing and reticulation in some retrospective analysis of 17 patients with advanced
patients.44 PLCH, echocardiography measurements of pulmonary
Avoidance of tobacco exposure, both active and passive, artery systolic pressure were > 35 mm Hg and
forms the cornerstone for treatment. Smoking cessation 50 mm Hg in 88% and 50% of patients, respectively.54
results in clinical improvement in approximately 20% to Presence of PH is also an independent prognostic factor
50% of patients.36 As in RB-ILD, there is paucity of data for increased mortality in PLCH. Extrapulmonary
about the efficacy of steroids. Data from previous studies involvement characterized by diabetes insipidus, bone
show that although up to 60% of patients treated with cysts, and skin lesions may be observed in approximately
corticosteroids had clinical improvement, 27% worsened 20% of the patients with PLCH.48
with this treatment.36 Because untreated patients may Plain chest radiographs may demonstrate upper lobe-
have deterioration of pulmonary function, predominant bilateral lung nodules, especially in the
corticosteroids and steroid-sparing immunosuppressant early stages of the disease, but are not diagnostic. HRCT
agents remain in use despite lack of concrete data. scans are superior to plain chest radiographs and
Prednisone is typically initiated at a dose of 0.5 to 1 mg/ typically reveal upper lobe nodules in early stages. These
kg and tapered over a few weeks to months, again based nodules later cavitate and ultimately form characteristic
on clinical response and tolerance. A single case report bizarre-shaped thick- and thin-walled cysts (Fig 3A). It
of clinical benefit with the use of macrolide therapy for is important to note that the nodules and cystic lesions
DIP has been reported.46 Therapy for nonsmoking- can co-exist with intervening normal lung parenchyma
related DIP should be tailored to the underlying in PLCH.52,55 In advanced disease, there may be
etiology, with an emphasis on avoidance of any form of evidence of extensive fibrosis and parenchymal
irritant exposure. Progressive disease warrants destruction. Thus, a combination of cystic and nodular
consideration for lung transplantation, if clinically lesions predominantly involving the upper lobes in a
appropriate, although recurrence has been reported.47 smoker strongly suggests PLCH. Costophrenic angles
Pulmonary Langerhans Cell Histiocytosis are characteristically spared.52 Areas of ground-glass
attenuation, which could represent other coexisting
PLCH is a nodular and cystic ILD seen typically in
smoking-related ILDs (RB-ILD/DIP), may also be
young adults in the age group of 20 to 40 years.48 There
observed.20
is no sex predilection. Smoking is the strongest risk
factor for development of this condition in adults and is A combination of smoking history with characteristic
reported in at least 90% of patients with adult PLCH. radiographic changes on HRCT scanning allows a
Although the precise mechanism of disease remains provisional diagnosis of PLCH. In cases in which
unclear, it is likely that PLCH represents a myeloid confirmation is needed, bronchoscopy with BAL and
neoplasm with inflammatory properties that is induced transbronchial biopsy may be useful. A BAL count of
by cigarette smoke in most instances. Cigarette smoke is Langerhans cells (CD1a stain-positive) > 5% may
likely the trigger that induces disease; however, the indicate PLCH but has poor sensitivity.56
identification of mutations affecting cell cycle pathways, Transbronchial biopsy has a diagnostic yield of 50%,
including the BRAF-V600E mutation (identified in up of whereas surgical biopsy results are usually definitive.56,57
50% of PLCH cases) and MAPK, have now shed light Light microscopy revealed granulomatous-like nodules
into novel mechanistic pathways by which this disease composed mainly of histiocytes that characteristically

400 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


Figure 3 – A, HRCT image of pulmonary Langerhans cell histiocytosis. HRCT scan of the chest of a 30-year-old female smoker demonstrates numerous
nodules and cysts, of variable size, shape, and wall thickness, predominantly in the mid and upper lungs. B, Histopathology of pulmonary Langerhans
cell histiocytosis. Nodular lesion containing numerous Langerhans cells well demonstrated by langerin immunohistochemical staining (inset). The
nodule may cavitate (not shown). Hematoxylin and eosin staining, 100 original magnification. See Figure 1 legend for expansion of abbreviation.

stain positive for CD1a antigen or langerin (Fig 3B). with RB-ILD and DIP (Table 1). Although the
However, concomitant severe PH and diffusion prevalence of SRIF-related changes in lungs range from
impairment may be prohibitive for pursuing lung 14% to 60% based on various small studies of pathology
biopsy. specimens, there is a clear increase in occurrence in
those who are heavy smokers.65-67 Both current and
Smoking cessation can stabilize lung function and
previous smokers are affected.
potentially improve physiological and radiographic
abnormalities in PLCH.58,59 For patients with refractory Although the series reported by Katzenstein et al67 did
or progressive PLCH, other therapies (including not have any distinct radiologic features, Yousem66
corticosteroids, immunosuppressants, and reported diffuse micronodules and GGOs, similar to RB-
chemotherapy) have been used, with some evidence of ILD. Although none of the studies had substantial long-
efficacy based on small series.59-61 Grobost et al62 also term follow-up (maximum follow up of 3 years), all the
reported improved outcomes (improved dyspnea and studies documented relative stability and only mild
increase in FEV1) in a series of patients with progressive progression, if any.66,67 Thus, the natural history of SRIF
PLCH who were treated with cladribine. PH may be may be more favorable in contrast to fibrotic lung
amenable to PH-specific therapies.63 However, none of diseases such as UIP or fibrotic nonspecific interstitial
the therapies for PLCH has been studied in a large-scale pneumonia.
randomized design. It remains to be seen whether
Due to a lack of well-defined clinical and radiologic
selective inhibitors of BRAF V600 kinase can be effective
features, SRIF will likely remain a histopathologic
in patients with PLCH who exhibit a BRAF-V600E
diagnosis. However, it should be considered in the
mutation. In advanced disease, lung transplantation
differential diagnosis of patients who present with
should be considered. Even with a 20% chance of
symptoms and radiography similar to RB-ILD and DIP.
recurrence, patients with PLCH tend to perform
On histopathology, it is characterized by hyalinized
satisfactorily following lung transplantation, with almost
deeply eosinophilic collagen with minimal surrounding
50% survival at 10 years following transplantation.64
inflammation (Fig 4). In contrast to UIP, there is
Smoking-Related Interstitial Fibrosis uniform, predominantly subpleural involvement with
little or no fibroblast foci. There is no temporal
SRIF is a relatively recently appreciated entity that has
heterogeneity. Honeycombing is rare in SRIF, whereas
gained prominence, mostly among pathologists,
emphysema and RB are almost always present.68 The
although it may have been described earlier using
typical pattern and greater degree of fibrosis in SRIF are
different terms such as “respiratory bronchiolitis with
also distinct from RB-ILD/DIP and thus can usually be
fibrosis” or “airspace enlargement with fibrosis.”65,66
distinguished. Collectively, these features allow SRIF to
Katzenstein et al67 described SRIF as a distinct form of
be distinguished from UIP, fibrotic nonspecific
chronic interstitial fibrosis characterized by uniform
interstitial pneumonia, and RB-ILD/DIP in most cases.
nonfocal deposition of hyalinized collagen in the
alveolar septal walls in smokers. However, its clinical Smoking cessation is plausibly the main treatment
ramifications are not well understood and likely overlap recommendation for these patients. Data on other

chestjournal.org 401
also present in a similar fashion and should be ruled out.
Other causes such as drug or toxin-mediated
eosinophilic processes may also involve the lung
parenchyma and should be considered.73,74
Plain chest radiographs demonstrate a pattern suggestive
of acute pulmonary edema with extensive airspace
opacities and pleural effusions in 50% of the patients.75
CT scans characteristically show bilateral diffuse GGOs
with no lobar predominance76 (Fig 5). Centrilobular
nodules and scattered airspace opacities may also be
Figure 4 – Histopathology of smoking-related interstitial fibrosis. Sub- rarely seen. Pleural effusion is frequently observed.70
pleural interstitial thickening due to prominent refractile collagen Results of pleural fluid studies typically reveal
deposition. Hematoxylin and eosin staining, 100 original
magnification. eosinophil-predominant exudates (22%-51% in one case
series).75 BAL typically reveals an increased eosinophil
count, with an average of 37% to 54%.70 Directing lavage
recommendations are lacking. However, as in RB-ILD/
to the area of maximal radiographic abnormality on the
DIP, a trial of corticosteroids is possibly worthwhile for
CT scan is more likely to demonstrate eosinophilia.
patients with significant disease. No studies have
Biopsy is rarely necessary, and the severe respiratory
documented the impact of immunosuppressant agents
failure, which affects many patients, precludes it.
or antifibrotic medications in patients with SRIF
However, when performed, the main biopsy findings
pathologic findings. include pronounced infiltration of eosinophils in the
The clinical significance of SRIF is unclear but possibly interstitium and alveolar spaces with features of diffuse
lies in the fact that it should be distinguished from other alveolar damage.70
forms of ILDs. Whether patients with SRIF will evolve to
Patients with AEP have a rapid and striking response to
develop honeycombing and UIP-like changes is unclear corticosteroids, with most patients exhibiting significant
at this time. However, based on data from available
clinical improvement within 24 to 48 h.77 Although the
studies, prognosis seems excellent.66,67
optimal dose of corticosteroids is uncertain, commonly
used dosages of IV methylprednisolone for patients with
Acute Eosinophilic Pneumonia
respiratory failure include 60 to 125 mg every 6 h until
AEP is characterized by the following: (1) febrile illness improvement, followed by initiation of oral prednisone,
of short duration (# 1 month, often < 1 week); (2) which is usually tapered off between 2 and 12 weeks.70
hypoxemic respiratory failure; (3) diffuse pulmonary Lower doses of oral prednisone and a shorter course
opacities on chest radiography; and (4) pulmonary may suffice in the treatment of those without respiratory
eosinophilia-bronchoscopy with BAL showing > failure.78 The tapering of steroids is tailored to the
25% eosinophils in the absence of parasitic infections, patient’s clinical response.
atopy, or asthma.18 The cause of AEP remains unknown
in many cases, and some investigators consider it to be
an acute hypersensitivity reaction to an unidentified
inhaled antigen.69 Of note, however, 40% of the patients
have a history of smoking cigarettes.70 Also, an
association between recent onset of cigarette smoking or
recent increase in the number of cigarettes has also been
consistently observed, suggesting a pathogenic role of
tobacco smoke constituents to trigger AEP.19,71
Smoking-related AEP is of greater disease severity and is
less likely to be associated with peripheral eosinophilia at
presentation compared with other forms of AEP.72 It is Figure 5 – HRCT image of acute eosinophilic pneumonia. HRCT image
also critical to consider other etiologies for similar of the chest of a 22-year-old male smoker who began smoking cigarettes
3 weeks earlier demonstrates patchy consolidative and ground-glass
presentation: fungal (eg, coccidioidomycosis) and opacities bilaterally along with pleural effusions. See Figure 1 legend for
parasitic infections (eg, ascariasis, strongyloidiasis) can expansion of abbreviation.

402 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


The prognosis of AEP is excellent, with complete Cumulative data from these studies show that even
radiographic resolution within 1 to 2 months following during a relatively short follow-up period (maximum of
initiation of corticosteroids.75,77 Relapse is uncommon 3 years in these studies), nonfibrotic interstitial lung
and is usually associated with resumption of cigarette changes may improve in approximately 50%, whereas
smoking.71 It is therefore crucial to recommend fibrotic interstitial changes are unlikely to regress and
avoidance of future cigarette smoking in these patients. can progress in up to 40% of patients. Another common
In a recently concluded study of patients with theme that emerged from these studies is that a greater
eosinophilic granulomatosis with polyangiitis, number of pack-years of smoking has an independent
mepolizumab (an anti-IL-5 monoclonal antibody that adjusted increased risk for causing both well-defined and
reduces blood eosinophil count) showed an ability to unclassifiable interstitial lung abnormalities in lungs of
sustain patients in remission for longer periods, thus smokers.82-84,86 Furthermore, the presence of these
allowing corticosteroid sparing.79 However, there is interstitial lung changes is also independently associated
currently no evidence to validate its use in patients with with increased all-cause mortality.87 The downstream
progressive or relapsing AEP. impact of these “subclinical” or “incidental” findings is
unclear; however, such findings are likely to increase the
“Subclinical” or “Incidental” Interstitial Lung need for imaging follow-up and downstream referrals to
Changes in Smokers an ILD specialist.
Varying degrees of interstitial lung abnormalities can be Although the economic repercussions of increased
seen in smokers and were often loosely referred to in the identification of these interstitial changes remain to be
past as the “dirty lung.”80,81 Although the entities studied, it offers the potential advantage of early
categorized under smoking-related ILDs constitute the diagnosis and an increased impetus for smoking
clinically apparent conditions, data from studies on cessation.88 The approach to these findings depends on
large cohorts of relatively healthy smokers suggest that the degree of respiratory impairment (symptoms and
smoking-related interstitial lung changes may be far pulmonary function abnormalities) and radiographic
more prevalent and of greater significance than pattern. In most situations, the identification of
previously recognized. Since the emergence of HRCT incidental smoking-related changes (ie, focal or diffuse
scans and also the increased implementation of lung GGOs and centrilobular nodules) provides an
cancer screening programs, various interstitial lung opportunity for emphasis on smoking cessation and
abnormalities, often incidental, are increasingly establishment of follow-up. In the symptomatic patient,
detected. Lederer et al82 reported an increase in high- pulmonary function testing assessment should be
attenuation areas (ground-glass and reticular considered. In individuals with progressive disease and
abnormalities) on CT scans in a population-based study fibrosis (ie, reticulation, traction bronchiectasis,
of relatively healthy smokers. The extent of high- honeycombing), further testing and management should
attenuation changes on the CT scan increased with be in line with the standard approach to fibrotic lung
increase in pack-years. diseases.
Based on studies that have spanned more than a decade,
the relative incidence of different types of interstitial Association Between Smoking and Other ILDs
lung changes seem to be variable. For instance, in a
study that included 692 participants of a lung cancer Idiopathic Pulmonary Fibrosis
screening program, RB-related changes were observed in Our current understanding of key pathways that trigger
15.7% of the patients, whereas “other chronic interstitial cigarette smoke-mediated fibrosis and stimulate its
pneumonia” and “indeterminate” interstitial changes progression in the lungs was described earlier in this
were noted in 3.8% and 3%, respectively.83 A review (the section entitled “Mechanisms and Spectrum
retrospective study in a lung cancer screening of Cigarette Smoke-Induced ILDs”). The prevalence of
population revealed some form of interstitial lung tobacco use in IPF ranges from 41% to 83%.89 Cigarette
abnormality in approximately 10% of their cohort.84 smoking has also been directly implicated in the
Washko et al85 reported interstitial changes in 8% of the pathogenesis of IPF.90 Studies in IPF suggest that
population of smokers who had an HRCT scan, and smoking is an independent risk factor with an OR of
those with interstitial changes had a reduced total lung 1.11 to 3.23 among ever smokers.91 Moreover, there is
capacity and lesser amount of emphysema. also an independent strong association between smoking

chestjournal.org 403
and the development of familial pulmonary fibrosis (OR: oxygen supplementation in hypoxemic patients, and
3.6 [95% CI: 1.3-9.4]).92 Although smoking may directly early referral for lung transplantation. Antifibrotic
promote fibrosis, it is likely that in IPF, cigarette smoke medications have not been explored in CPFE, and their
and other environmental or endogenous profibrotic effects on disease progression are unknown.
stimuli synergize to promote interstitial fibrosis in
predisposed individuals. Furthermore, cigarette smoking Connective Tissue Disease-Associated ILD
influences the clinical course of IPF. In a study by
The impact of cigarette smoking in various types of
Antoniou et al93 of 249 patients with IPF, severity CTD-ILD is not clear. Among the various autoimmune
adjusted survival was higher among nonsmokers
diseases, the association with cigarette smoking is most
compared with current or ever smokers. In addition, established for RA.102,103 Several epidemiologic studies
smoking is also a risk factor for development of acute
have shown that the risk of developing seropositive RA
exacerbation of IPF. Although these data were recently
is higher among those with tobacco exposure.102-104
contradicted, the conflicting results are likely due to
Longer duration and severity of smoking has been
limitations of lead time bias (early diagnosis in smokers)
shown to increase the risk for RA (approximate relative
and the retrospective nature of some of these studies.94,95
risk of 1.5 times), and the risk remains high even after
Combined Pulmonary Fibrosis and Emphysema smoking cessation.104 Klareskog et al105 reported a
major gene-environment interaction between smoking
The association between emphysema and cigarette and HLA-DR shared epitope (HLA-DR SE) genes that
smoking is well established. However, when emphysema were evident for seropositive RA. In this study, the
and pulmonary fibrosis co-exist in the same patient, it combination of smoking history and the presence of
represents a unique syndrome called CPFE.96 CPFE is double copies of HLA-DR SE genes increased the risk
characterized by upper lobe emphysema and lower lobe for seropositive RA 21-fold compared with the risk
interstitial fibrosis. However, these patients have a among nonsmokers. However, specific pathogenetic
different natural history than patients with either interactions by which smoking and other factors
pulmonary fibrosis or emphysema alone.97 increase the risk of RA and ILD are not well understood.
A consensus definition for CPFE does not exist. In the
RA is associated with autoantibodies to various self-
broadest sense, this term should include all patients with
antigens, including citrullinated proteins such as type II
co-existent emphysema and fibrosis on HRCT
collagen, which is found in the synovial tissues and
scanning.97 Most of the affected patients tend to be male
lungs. Smoking increases the production of citrulline-
smokers (with > 40 pack-year smoking history), age >
modified proteins secondary to an increase in
65 years, and severely symptomatic.98 Because
peptidylarginine deiminase enzymes that are involved
emphysema is associated with increased lung volumes
in the citrullination of native proteins.105 Therefore, in
and compliance and pulmonary fibrosis with opposite
predisposed individuals, an enhanced production of
physiological effects, pulmonary function test results
anti-cyclic citrullinated peptide antibodies is promoted
characteristically show normal or near-normal
by cigarette smoke exposure. How this scenario in turn
spirometry values and preserved lung volumes with
leads to enhanced pulmonary inflammation of lung
severely reduced diffusing capacity of the lung for
fibrosis is not yet known.
carbon monoxide.97
Moreover, cigarette smoking has an impact on response
Mortality in CPFE is significant, with a median reported
to treatment in RA, with smokers typically exhibiting a
survival of 2.1 to 8.5 years, with an increased incidence
poor response to methotrexate and other biologic
of PH and lung cancer in this group.97,99 The presence of
disease-modifying antirheumatic drugs compared with
PH portends a poor prognosis, with predicted 1-year
never smokers.106 Hudson et al107 evaluated the effects
survival rate of 60% in patients with CPFE and PH.99
of cigarette smoking in a cohort of patients with
The prevalence of PH is higher than that reported in IPF
systemic sclerosis and found a strong negative
or emphysema.96,97,99 Although the initial studies did
correlation between smoking intensity and spirometry,
not identify a mortality difference between patients with
as well as a trend toward worsening diffusing capacity of
CPFE and non-CPFE IPF, a recent analysis showed a
the lung for carbon monoxide.
distinctly lower 5-year survival and higher mortality
compared with IPF.100,101 Treatment options are CPFE has also been described within the context of
unfortunately limited and include smoking cessation, smokers with CTD-ILDs. Patients were younger, more

404 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


frequently female, and tended to present with a less Acknowledgments
severe prognosis.108 The physiological profile was Financial/nonfinancial disclosure: None declared.
similar with idiopathic CPFE, but PH was less severe,
raising the hypothesis that the better prognosis of CTD- References
ILDs delays the appearance and blunts the severity of 1. Zanetti F, Giacomello M, Donati Y, Carnesecchi S, Frieden M,
Barazzone-Argiroffo C. Nicotine mediates oxidative stress and
PH in CTD-related CPFE.108 apoptosis through cross talk between NOX1 and Bcl-2 in lung
epithelial cells. Free Radic Biol Med. 2014;76:173-184.
Interestingly, a recent study found that current tobacco 2. Stevenson CS, Docx C, Webster R, et al. Comprehensive gene
smoking, although of relatively low incidence among expression profiling of rat lung reveals distinct acute and chronic
responses to cigarette smoke inhalation. Am J Physiol Lung Cell
patients in the study cohort, has a negative association in Mol Physiol. 2007;293(5):L1183-L1193.
patients with Sjögren syndrome, as smoking seems to 3. Chen R, Zhang K, Chen H, et al. Telomerase deficiency causes
alveolar stem cell senescence-associated low-grade inflammation in
delay progression of disease-associated humoral and lungs. J Biol Chem. 2015;290(52):30813-30829.
cellular autoimmunity.109 Despite the protective effects 4. Alder JK, Guo N, Kembou F, et al. Telomere length is a
of tobacco smoking on Sjögren syndrome determinant of emphysema susceptibility. Am J Respir Crit Care
Med. 2011;184(8):904-912.
manifestations, it is still associated with lung disease,
5. Alder JK, Chen JJ, Lancaster L, et al. Short telomeres are a risk
cardiovascular risk, and malignancy, and should thus be factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci U S A.
discouraged. 2008;105(35):13051-13056.
6. Schafer MJ, White TA, Iijima K, et al. Cellular senescence mediates
fibrotic pulmonary disease. Nat Commun. 2017;8:14532.
7. Churg A, Zhou S, Preobrazhenska O, Tai H, Wang R, Wright JL.
Conclusions Expression of profibrotic mediators in small airways versus
Cigarette smoking results in manifold changes to the parenchyma after cigarette smoke exposure. Am J Respir Cell Mol
Biol. 2009;40(3):268-276.
airways and lung parenchyma. Aside from the
8. Checa M, Hagood JS, Velazquez-Cruz R, et al. Cigarette smoke
destruction to alveolar walls that results in emphysema, enhances the expression of profibrotic molecules in alveolar
tobacco smoke is also a well-known trigger for varying epithelial cells. PLoS One. 2016;11(3):e0150383.
9. Daniels CE, Wilkes MC, Edens M, et al. Imatinib mesylate inhibits
degrees of interstitial changes in the lungs that manifest the profibrogenic activity of TGF-beta and prevents bleomycin-
in diverse ways in the affected patient population. mediated lung fibrosis. J Clin Invest. 2004;114(9):1308-1316.
Clinically apparent smoking-related ILDs fall into a 10. Asakura S, Colby TV, Limper AH. Tissue localization of
transforming growth factor-beta1 in pulmonary eosinophilic
spectrum of disorders that likely have a causal granuloma. Am J Respir Crit Care Med. 1996;154(5):1525-1530.
relationship with tobacco use and include RB-ILD, DIP, 11. Uebelhoer M, Bewig B, Kreipe H, Nowak D, Magnussen H, Barth J.
a subset of AEP, and PLCH. The fact that cigarette Modulation of fibroblast activity in histiocytosis X by platelet-
derived growth factor. Chest. 1995;107(3):701-705.
smoke promotes a profibrotic environment through
12. Prasse A, Stahl M, Schulz G, et al. Essential role of osteopontin in
certain endogenous and exogenous growth factors also smoking-related interstitial lung diseases. Am J Pathol. 2009;174(5):
lends support to its pathogenetic role in conditions such 1683-1691.

as IPF and CPFE. 13. Shan M, Yuan X, Song LZ, et al. Cigarette smoke induction of
osteopontin (SPP1) mediates T(H)17 inflammation in human and
experimental emphysema. Sci Transl Med. 2012;4(117):117ra9.
Our understanding of the spectrum of smoking-related
14. Tazi A, Bonay M, Bergeron A, Grandsaigne M, Hance AJ, Soler P.
interstitial lung changes continues to evolve with Role of granulocyte-macrophage colony stimulating factor (GM-
recognition of novel entities such as SRIF, which despite CSF) in the pathogenesis of adult pulmonary histiocytosis X.
Thorax. 1996;51(6):611-614.
distinct histopathologic features, remains of unclear 15. Vassallo R, Walters PR, Lamont J, Kottom TJ, Yi ES, Limper AH.
clinical relevance. We also emphasize the relevance of Cigarette smoke promotes dendritic cell accumulation in COPD; a
Lung Tissue Research Consortium study. Respir Res. 2010;11:45.
appreciating “subclinical” or “incidental” interstitial
16. Jhun BW, Kim SJ, Kim K, Lee JE, Hong DJ. Clinical implications of
changes in relatively healthy smokers. Increased correlation between peripheral eosinophil count and serum levels
recognition of these changes on CT scans obtained for of IL-5 and tryptase in acute eosinophilic pneumonia. Respir Med.
2014;108(11):1655-1662.
other reasons (especially in a high-risk population such
17. Mato N, Bando M, Kusano A, et al. Clinical significance of
as patients enrolled in a lung cancer screening program) interleukin 33 (IL-33) in patients with eosinophilic pneumonia.
will be a source of an increased number of referrals to Allergol Int. 2013;62(1):45-52.
ILD specialists. Regardless of the type of ILD, smoking 18. Allen JN, Pacht ER, Gadek JE, Davis WB. Acute eosinophilic
pneumonia as a reversible cause of noninfectious respiratory
cessation should continue to be the principal failure. N Engl J Med. 1989;321(9):569-574.
recommendation for patients whose diagnosis falls into 19. Shorr AF, Scoville SL, Cersovsky SB, et al. Acute eosinophilic
this category of lung diseases while our knowledge of pneumonia among US military personnel deployed in or near Iraq.
JAMA. 2004;292(24):2997-3005.
the role of other potential therapies continues to 20. Vassallo R, Jensen EA, Colby TV, et al. The overlap between
expand. respiratory bronchiolitis and desquamative interstitial pneumonia

chestjournal.org 405
in pulmonary Langerhans cell histiocytosis: high-resolution CT, 41. Abraham JL, Hertzberg MA. Inorganic particulates associated with
histologic, and functional correlations. Chest. 2003;124(4): desquamative interstitial pneumonia. Chest. 1981;80(suppl 1):
1199-1205. 67-70.
21. Niewoehner DE, Kleinerman J, Rice DB. Pathologic changes in the 42. Flores-Franco RA, Luevano-Flores E, Gaston-Ramirez C.
peripheral airways of young cigarette smokers. N Engl J Med. Sirolimus-associated desquamative interstitial pneumonia.
1974;291(15):755-758. Respiration. 2007;74(2):237-238.
22. Myers JL, Veal CF Jr, Shin MS, Katzenstein AL. Respiratory 43. Iskandar SB, McKinney LA, Shah L, Roy TM, Byrd RP Jr.
bronchiolitis causing interstitial lung disease. A clinicopathologic Desquamative interstitial pneumonia and hepatitis C virus
study of six cases. Am Rev Respir Dis. 1987;135(4):880-884. infection: a rare association. South Med J. 2004;97(9):890-893.
23. Ryu JH, Myers JL, Capizzi SA, Douglas WW, Vassallo R, 44. Kawabata Y, Takemura T, Hebisawa A, et al. Desquamative
Decker PA. Desquamative interstitial pneumonia and respiratory interstitial pneumonia may progress to lung fibrosis as
bronchiolitis-associated interstitial lung disease. Chest. 2005;127(1): characterized radiologically. Respirology. 2012;17(8):1214-1221.
178-184.
45. Kawabata Y, Takemura T, Hebisawa A, et al. Eosinophilia in
24. Moon J, du Bois RM, Colby TV, Hansell DM, Nicholson AG. bronchoalveolar lavage fluid and architectural destruction are
Clinical significance of respiratory bronchiolitis on open lung features of desquamative interstitial pneumonia. Histopathology.
biopsy and its relationship to smoking related interstitial lung 2008;52(2):194-202.
disease. Thorax. 1999;54(11):1009-1014.
46. Knyazhitskiy A, Masson RG, Corkey R, Joiner J. Beneficial response
25. Park JS, Brown KK, Tuder RM, Hale VA, King TE Jr, Lynch DA. to macrolide antibiotic in a patient with desquamative interstitial
Respiratory bronchiolitis-associated interstitial lung disease: pneumonia refractory to corticosteroid therapy. Chest. 2008;134(1):
radiologic features with clinical and pathologic correlation. 185-187.
J Comput Assist Tomogr. 2002;26(1):13-20.
47. Verleden GM, Sels F, Van Raemdonck D, Verbeken EK, Lerut T,
26. Cottin V, Streichenberger N, Gamondes JP, Thevenet F, Loire R, Demedts M. Possible recurrence of desquamative interstitial
Cordier JF. Respiratory bronchiolitis in smokers with spontaneous pneumonitis in a single lung transplant recipient. Eur Respir J.
pneumothorax. Eur Respir J. 1998;12(3):702-704. 1998;11(4):971-974.
27. Fraig M, Shreesha U, Savici D, Katzenstein AL. Respiratory
48. Vassallo R, Ryu JH, Schroeder DR, Decker PA, Limper AH. Clinical
bronchiolitis: a clinicopathologic study in current smokers, ex-
outcomes of pulmonary Langerhans’-cell histiocytosis in adults.
smokers, and never-smokers. Am J Surg Pathol. 2002;26(5):
N Engl J Med. 2002;346(7):484-490.
647-653.
28. Elkin SL, Nicholson AG, du Bois RM. Desquamative interstitial 49. Roden AC, Hu X, Kip S, et al. BRAF V600E expression in
pneumonia and respiratory bronchiolitis-associated interstitial lung Langerhans cell histiocytosis: clinical and immunohistochemical
disease. Semin Respir Crit Care Med. 2001;22(4):387-398. study on 25 pulmonary and 54 extrapulmonary cases. Am J Surg
Pathol. 2014;38(4):548-551.
29. Hartman TE, Primack SL, Swensen SJ, Hansell D, McGuinness G,
Muller NL. Desquamative interstitial pneumonia: thin-section CT 50. Kamionek M, Ahmadi Moghaddam P, Sakhdari A, et al. Mutually
findings in 22 patients. Radiology. 1993;187(3):787-790. exclusive extracellular signal-regulated kinase pathway mutations
are present in different stages of multi-focal pulmonary Langerhans
30. Godbert B, Wissler MP, Vignaud JM. Desquamative interstitial cell histiocytosis supporting clonal nature of the disease.
pneumonia: an analytic review with an emphasis on aetiology. Eur Histopathology. 2016;69(3):499-509.
Respir Rev. 2013;22(128):117-123.
51. Mendez JL, Nadrous HF, Vassallo R, Decker PA, Ryu JH.
31. Yousem SA, Colby TV, Gaensler EA. Respiratory bronchiolitis- Pneumothorax in pulmonary Langerhans cell histiocytosis. Chest.
associated interstitial lung disease and its relationship to 2004;125(3):1028-1032.
desquamative interstitial pneumonia. Mayo Clin Proc. 1989;64(11):
1373-1380. 52. Abbott GF, Rosado-de-Christenson ML, Franks TJ, Frazier AA,
Galvin JR. From the archives of the AFIP: pulmonary Langerhans
32. Craig PJ, Wells AU, Doffman S, et al. Desquamative interstitial cell histiocytosis. Radiographics. 2004;24(3):821-841.
pneumonia, respiratory bronchiolitis and their relationship to
smoking. Histopathology. 2004;45(3):275-282. 53. Fartoukh M, Humbert M, Capron F, et al. Severe pulmonary
hypertension in histiocytosis X. Am J Respir Crit Care Med.
33. Churg A, Muller NL, Wright JL. Respiratory bronchiolitis/
2000;161(1):216-223.
interstitial lung disease: fibrosis, pulmonary function, and evolving
concepts. Arch Pathol Lab Med. 2010;134(1):27-32. 54. Chaowalit N, Pellikka PA, Decker PA, et al. Echocardiographic and
34. Portnoy J, Veraldi KL, Schwarz MI, et al. Respiratory bronchiolitis- clinical characteristics of pulmonary hypertension complicating
interstitial lung disease: long-term outcome. Chest. 2007;131(3): pulmonary Langerhans cell histiocytosis. Mayo Clin Proc.
664-671. 2004;79(10):1269-1275.

35. Liebow AA, Steer A, Billingsley JG. Desquamative interstitial 55. Kim HJ, Lee KS, Johkoh T, et al. Pulmonary Langerhans cell
pneumonia. Am J Med. 1965;39:369-404. histiocytosis in adults: high-resolution CT-pathology comparisons
and evolutional changes at CT. Eur Radiol. 2011;21(7):1406-1415.
36. Carrington CB, Gaensler EA, Coutu RE, FitzGerald MX, Gupta RG.
Natural history and treated course of usual and desquamative 56. Harari S, Torre O, Cassandro R, Taveira-DaSilva AM, Moss J.
interstitial pneumonia. N Engl J Med. 1978;298(15):801-809. Bronchoscopic diagnosis of Langerhans cell histiocytosis and
lymphangioleiomyomatosis. Respir Med. 2012;106(9):1286-1292.
37. Ishii H, Iwata A, Sakamoto N, Mizunoe S, Mukae H, Kadota J.
Desquamative interstitial pneumonia (DIP) in a patient with 57. Baqir M, Vassallo R, Maldonado F, Yi ES, Ryu JH. Utility of
rheumatoid arthritis: is DIP associated with autoimmune bronchoscopy in pulmonary Langerhans cell histiocytosis.
disorders? Intern Med. 2009;48(10):827-830. J Bronchology Interv Pulmonol. 2013;20(4):309-312.
38. Swartz JS, Chatterjee S, Parambil JG. Desquamative interstitial 58. Negrin-Dastis S, Butenda D, Dorzee J, Fastrez J, d’Odemont JP.
pneumonia as the initial manifestation of systemic sclerosis. J Clin Complete disappearance of lung abnormalities on high-resolution
Rheumatol. 2010;16(6):284-286. computed tomography: a case of histiocytosis X. Can Respir J.
2007;14(4):235-237.
39. Lougheed MD, Roos JO, Waddell WR, Munt PW. Desquamative
interstitial pneumonitis and diffuse alveolar damage in textile 59. Elia D, Torre O, Cassandro R, Caminati A, Harari S. Pulmonary
workers. Potential role of mycotoxins. Chest. 1995;108(5): Langerhans cell histiocytosis: a comprehensive analysis of 40
1196-1200. patients and literature review. Eur J Intern Med. 2015;26(5):
351-356.
40. Dvorackova I, Pichova V. Pulmonary interstitial fibrosis with
evidence of aflatoxin B1 in lung tissue. J Toxicol Environ Health. 60. Lazor R, Etienne-Mastroianni B, Khouatra C, Tazi A, Cottin V,
1986;18(1):153-157. Cordier JF. Progressive diffuse pulmonary Langerhans cell

406 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]


histiocytosis improved by cladribine chemotherapy. Thorax. 82. Lederer DJ, Enright PL, Kawut SM, et al. Cigarette smoking is
2009;64(3):274-275. associated with subclinical parenchymal lung disease: the Multi-
Ethnic Study of Atherosclerosis (MESA)-Lung study. Am J Respir
61. Mason RH, Foley NM, Branley HM, et al. Pulmonary Langerhans
Crit Care Med. 2009;180(5):407-414.
cell histiocytosis (PLCH): a new UK register. Thorax. 2014;69(8):
766-767. 83. Sverzellati N, Guerci L, Randi G, et al. Interstitial lung diseases in a
lung cancer screening trial. Eur Respir J. 2011;38(2):392-400.
62. Grobost V, Khouatra C, Lazor R, Cordier JF, Cottin V.
Effectiveness of cladribine therapy in patients with pulmonary 84. Jin GY, Lynch D, Chawla A, et al. Interstitial lung abnormalities in
Langerhans cell histiocytosis. Orphanet J Rare Dis. 2014;9:191. a CT lung cancer screening population: prevalence and progression
rate. Radiology. 2013;268(2):563-571.
63. Le Pavec J, Lorillon G, Jais X, et al. Pulmonary Langerhans cell
histiocytosis-associated pulmonary hypertension: clinical 85. Washko GR, Hunninghake GM, Fernandez IE, et al. Lung volumes
characteristics and impact of pulmonary arterial hypertension and emphysema in smokers with interstitial lung abnormalities.
therapies. Chest. 2012;142(5):1150-1157. N Engl J Med. 2011;364(10):897-906.
64. Dauriat G, Mal H, Thabut G, et al. Lung transplantation for 86. Salvatore M, Henschke CI, Yip R, et al. Journal Club: evidence of
pulmonary langerhans’ cell histiocytosis: a multicenter analysis. interstitial lung disease on low-dose chest CT images: prevalence,
Transplantation. 2006;81(5):746-750. patterns, and progression. AJR Am J Roentgenol. 2016;206(3):
487-494.
65. Kawabata Y, Hoshi E, Murai K, et al. Smoking-related changes in
the background lung of specimens resected for lung cancer: a 87. Putman RK, Hatabu H, Araki T, et al. Association between
semiquantitative study with correlation to postoperative course. interstitial lung abnormalities and all-cause mortality. JAMA.
Histopathology. 2008;53(6):707-714. 2016;315(7):672-681.
66. Yousem SA. Respiratory bronchiolitis-associated interstitial lung 88. Borondy Kitts AK, McKee AB, Regis SM, Wald C, Flacke S,
disease with fibrosis is a lesion distinct from fibrotic nonspecific McKee BJ. Smoking cessation results in a clinical lung cancer
interstitial pneumonia: a proposal. Mod Pathol. 2006;19(11): screening program. J Thorac Dis. 2016;8(suppl 6):S481-S487.
1474-1479.
89. Oh CK, Murray LA, Molfino NA. Smoking and idiopathic
67. Katzenstein AL, Mukhopadhyay S, Zanardi C, Dexter E. Clinically pulmonary fibrosis. Pulm Med. 2012;2012:808260.
occult interstitial fibrosis in smokers: classification and significance
90. Baumgartner KB, Samet JM, Stidley CA, Colby TV, Waldron JA.
of a surprisingly common finding in lobectomy specimens. Hum
Cigarette smoking: a risk factor for idiopathic pulmonary fibrosis.
Pathol. 2010;41(3):316-325.
Am J Respir Crit Care Med. 1997;155(1):242-248.
68. Katzenstein AL. Smoking-related interstitial fibrosis (SRIF),
91. Taskar V, Coultas D. Exposures and idiopathic lung disease. Semin
pathogenesis and treatment of usual interstitial pneumonia (UIP),
Respir Crit Care Med. 2008;29(6):670-679.
and transbronchial biopsy in UIP. Mod Pathol. 2012;25(suppl 1):
S68-S78. 92. Steele MP, Speer MC, Loyd JE, et al. Clinical and pathologic
features of familial interstitial pneumonia. Am J Respir Crit Care
69. Badesch DB, King TE Jr, Schwarz MI. Acute eosinophilic
Med. 2005;172(9):1146-1152.
pneumonia: a hypersensitivity phenomenon? Am Rev Respir Dis.
1989;139(1):249-252. 93. Antoniou KM, Hansell DM, Rubens MB, et al. Idiopathic
pulmonary fibrosis: outcome in relation to smoking status. Am J
70. Bhatt NY, Allen JN. Update on eosinophilic lung diseases. Semin
Respir Crit Care Med. 2008;177(2):190-194.
Respir Crit Care Med. 2012;33(5):555-571.
94. Collard HR, Richeldi L, Kim DS, et al. Acute exacerbations in the
71. Uchiyama H, Suda T, Nakamura Y, et al. Alterations in smoking
INPULSIS trials of nintedanib in idiopathic pulmonary fibrosis.
habits are associated with acute eosinophilic pneumonia. Chest.
Eur Respir J. 2017;49(5).
2008;133(5):1174-1180.
95. Kishaba T, Nagano H, Nei Y, Yamashiro S. Clinical characteristics
72. De Giacomi F, Decker PA, Vassallo R, Ryu JH. Acute eosinophilic
of idiopathic pulmonary fibrosis patients according to their
pneumonia: correlation of clinical characteristics with underlying
smoking status. J Thorac Dis. 2016;8(6):1112-1120.
cause. Chest. 2017;152(2):379-385.
96. Cottin V, Nunes H, Brillet PY, et al. Combined pulmonary fibrosis
73. Lepine PA, Dumas A, Boulet LP. Pulmonary eosinophilia from
and emphysema: a distinct underrecognised entity. Eur Respir J.
inhaled colistin. Chest. 2017;151(1):e1-e3.
2005;26(4):586-593.
74. Miller BA, Gray A, Leblanc TW, Sexton DJ, Martin AR, Slama TG.
97. Jankowich MD, Rounds SI. Combined pulmonary fibrosis and
Acute eosinophilic pneumonia secondary to daptomycin: a report
emphysema syndrome: a review. Chest. 2012;141(1):222-231.
of three cases. Clin Infect Dis. 2010;50(11):e63-e68.
98. Portillo K, Morera J. Combined pulmonary fibrosis and
75. Philit F, Etienne-Mastroianni B, Parrot A, Guerin C, Robert D,
emphysema syndrome: a new phenotype within the spectrum of
Cordier JF. Idiopathic acute eosinophilic pneumonia: a study of 22
smoking-related interstitial lung disease. Pulm Med. 2012;2012:
patients. Am J Respir Crit Care Med. 2002;166(9):1235-1239.
867870.
76. Daimon T, Johkoh T, Sumikawa H, et al. Acute eosinophilic
99. Cottin V, Le Pavec J, Prevot G, et al. Pulmonary hypertension in
pneumonia: thin-section CT findings in 29 patients. Eur J Radiol.
patients with combined pulmonary fibrosis and emphysema
2008;65(3):462-467.
syndrome. Eur Respir J. 2010;35(1):105-111.
77. Rhee CK, Min KH, Yim NY, et al. Clinical characteristics and
100. Ryerson CJ, Hartman T, Elicker BM, et al. Clinical features and
corticosteroid treatment of acute eosinophilic pneumonia. Eur
outcomes in combined pulmonary fibrosis and emphysema in
Respir J. 2013;41(2):402-409.
idiopathic pulmonary fibrosis. Chest. 2013;144(1):234-240.
78. Jhun BW, Kim SJ, Kim K, Lee JE. Outcomes of rapid corticosteroid
101. Zhang L, Zhang C, Dong F, et al. Combined pulmonary fibrosis and
tapering in acute eosinophilic pneumonia patients with initial
emphysema: a retrospective analysis of clinical characteristics,
eosinophilia. Respirology. 2015;20(8):1241-1247.
treatment and prognosis. BMC Pulm Med. 2016;16(1):137.
79. Wechsler ME, Akuthota P, Jayne D, et al. Mepolizumab or placebo
102. Stolt P, Bengtsson C, Nordmark B, et al. Quantification of the
for eosinophilic granulomatosis with polyangiitis. N Engl J Med.
influence of cigarette smoking on rheumatoid arthritis: results from
2017;376(20):1921-1932.
a population based case-control study, using incident cases. Ann
80. Guckel C, Hansell DM. Imaging the ’dirty lung’—has high Rheum Dis. 2003;62(9):835-841.
resolution computed tomography cleared the smoke? Clin Radiol.
103. Criswell LA, Merlino LA, Cerhan JR, et al. Cigarette smoking and
1998;53(10):717-722.
the risk of rheumatoid arthritis among postmenopausal women:
81. King TE Jr. Smoking and subclinical interstitial lung disease. N Engl results from the Iowa Women’s Health Study. Am J Med.
J Med. 2011;364(10):968-970. 2002;112(6):465-471.

chestjournal.org 407
104. Costenbader KH, Feskanich D, Mandl LA, Karlson EW. Smoking 107. Hudson M, Lo E, Lu Y, et al. Cigarette smoking in patients with
intensity, duration, and cessation, and the risk of rheumatoid systemic sclerosis. Arthritis Rheum. 2011;63(1):230-238.
arthritis in women. Am J Med. 2006;119(6):503.e1-503.e9.
108. Cottin V, Nunes H, Mouthon L, et al. Combined pulmonary
105. Klareskog L, Stolt P, Lundberg K, et al. A new model for an etiology fibrosis and emphysema syndrome in connective tissue disease.
of rheumatoid arthritis: smoking may trigger HLA-DR (shared Arthritis Rheum. 2011;63(1):295-304.
epitope)-restricted immune reactions to autoantigens modified by
citrullination. Arthritis Rheum. 2006;54(1):38-46. 109. Stone DU, Fife D, Brown M, et al. Effect of tobacco smoking
106. Samara KD, Margaritopoulos G, Wells AU, Siafakas NM, on the clinical, histopathological, and serological
Antoniou KM. Smoking and pulmonary fibrosis: novel insights. manifestations of Sjogren’s syndrome. PLoS One. 2017;12(2):
Pulm Med. 2011;2011:461439. e0170249.

408 Recent Advances in Chest Medicine [ 154#2 CHEST AUGUST 2018 ]

You might also like