You are on page 1of 11

269

Pulmonary Langerhans Cell Histiocytosis


Brian Shaw, MD1 Michael Borchers, PhD2,3 Dani Zander, MD4 Nishant Gupta, MD, MS2,3

1 Department of Internal Medicine, University of Cincinnati, Address for correspondence Nishant Gupta, MD, MS, Division of
Cincinnati, Ohio Pulmonary, Critical Care, and Sleep Medicine, University of Cincinnati,
2 Medical Service, Veterans Affairs Medical Center, Cincinnati, Ohio 231 Albert Sabin Way, MSB Room 6053, ML 0564, Cincinnati, OH
3 Division of Pulmonary, Critical Care, and Sleep Medicine, University 45267 (e-mail: guptans@ucmail.uc.edu).
of Cincinnati, Cincinnati, Ohio
4 Department of Pathology and Laboratory Medicine, University of
Cincinnati, Cincinnati, Ohio

Semin Respir Crit Care Med 2020;41:269–279.

Abstract Pulmonary Langerhans cell histiocytosis (PLCH) is a diffuse cystic lung disease that is
strongly associated with exposure to cigarette smoke. Recently, activating pathogenic

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


mutations in the mitogen-activated protein kinase pathway have been described in the
dendritic cells in patients with PLCH and have firmly established PLCH to be an
inflammatory myeloid neoplasm. Disease course and prognosis in PLCH are highly
variable among individual patients, ranging from spontaneous resolution to develop-
ment of pulmonary hypertension and progression to terminal respiratory failure. A
subset of patients with PLCH may have extrapulmonary involvement, typically involv-
Keywords ing the skeletal system in the form of lytic lesions, skin lesions, or the central nervous
► PLCH system most commonly manifesting in the form of diabetes insipidus. Smoking
► pneumothorax cessation is the cornerstone of treatment in patients with PLCH and can lead to
► MAPK disease regression or stabilization in a substantial proportion of patients. Further
► BRAF insight into the underlying molecular pathogenesis of PLCH has paved the way for the
► cladribine future development of disease-specific biomarkers and targeted treatment options
► smoking directed against the central disease-driving mutations.

Pulmonary Langerhans cell histiocytosis (PLCH) is a rare cytoses), the R group (Rosai–Dorfman’s disease and miscella-
diffuse cystic lung disease characterized by the development neous noncutaneous, non-Langerhans cell histiocytoses), and
of centrilobular lesions composed of CD1aþ dendritic cells the H group (hemophagocytic, lymphohistiocytic, and macro-
(DCs) and other inflammatory cells, with variable degrees of phage activation syndrome). The L group contains several
fibrosis. These DCs have recently been discovered to carry a diseases: Langerhans cell histiocytosis (LCH), indeterminant
variety of damaging mutations that lead to constitutive cell histiocytosis, Erdheim–Chester’s disease (ECD), and mixed
activation of the mitogen-activated protein kinase (MAPK) LCH/ECD diseases. LCH is further categorized on the basis
pathway. As such, PLCH is now classified as an inflammatory of organ or system involvement: single system LCH, LCH with
myeloid neoplasm. In addition to the MAPK mutations, PLCH pulmonary involvement (PLCH), multisystem LCH with
has a strong association with cigarette smoke exposure and is involvement of risk organs, multisystem LCH without involve-
almost exclusively seen in active/former smokers. ment of risk organs (risk organs being liver, spleen, and
PLCH is part of a spectrum of histiocytic disorders that was bone marrow), and LCH associated with another myeloprolif-
first described by Farber in 1941.1 The most recent classification erative/myelodysplastic disorder.2
scheme for histiocytic disorders recognizes the following five The majority of adults with PLCH tend to have isolated
groups: the L group (Langerhans), the C group (cutaneous and pulmonary disease; however, extrapulmonary involvement
mucocutaneous histiocytosis), the M group (malignant histio- can be seen in 15 to 20% of cases.3–5 The most common

Issue Theme Orphan Lung Diseases; Copyright © 2020 by Thieme Medical DOI https://doi.org/
Guest Editors: Jay H. Ryu, MD, Nishant Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0039-1700996.
Gupta, MD New York, NY 10001, USA. ISSN 1069-3424.
Tel: +1(212) 760-0888.
270 Pulmonary Langerhans Cell Histiocytosis Shaw et al.

extrapulmonary features are lytic bone lesions, diabetes


insipidus from posterior pituitary involvement, and skin
lesions.3,5–12 Although involvement of risk organs (liver,
spleen, and hematopoietic system) is rare, it portends a
poor prognosis.13 The course of PLCH is variable and unpre-
dictable, ranging from asymptomatic with spontaneous res-
olution to relentless progression despite smoking cessation
and aggressive pharmacological treatment.

Epidemiology
Due to its rarity, limited epidemiological data are available
for PLCH. There is a strong association with exposure to
cigarette (and marijuana) smoke and the development of
PLCH.14 PLCH can occur at any age; however, it is most
commonly diagnosed in patients aged 20 to 40 years.1,4,15
There is no known sex predilection, and both males and
females appear to be equally affected by PLCH. There was a

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


suggestion of male predominance in the past, however, that
likely relates to the differences in historical smoking rates
between males and females.16 It is estimated that PLCH
accounts for 3 to 5% of all diffuse lung diseases in
adults.17–19 A national survey of hospital discharges con-
ducted in Japan estimated the prevalence of PLCH to be 0.27
per 100,000 males and 0.07 per 100,000 females in the
Japanese population.12 Extrapolating these numbers to the
current population estimates suggests that there are 1,100
patients with PLCH in the United States and 26,000 world-
wide. Similar to other rare diseases, these numbers are
almost certainly an underestimate of the true disease
prevalence.

Pathogenesis
The pathogenesis of PLCH is now better understood and can
broadly be visualized as progression along a linear pathway:
DCs harboring activating MAPK mutations accumulate in the
pulmonary parenchyma under the influence of cigarette
smoke, where they lead to secondary immune system activa-
tion and migration, forming cellular nodules. The accumulated
leukocytes including myeloid cells, monocytes, and cytotoxic
lymphocytes20 release cytotoxic mediators and matrix-degrad-
ing enzymes which contribute to the bronchiolar destruction
characteristic of PLCH (►Fig. 1).

Accumulation of Dendritic Cells


DCs are members of the mononuclear phagocyte system and
are composed of a heterogenous population of cells whose
primary function is to serve as antigen-presenting cells to T-
cells. Langerhans cells (LCs) are a subset of DCs that are localized Fig. 1 The pathogenesis of PLCH. DC, dendritic cells; MAPK, mitogen-
to the epidermis and mucosal epithelium (including the tra- activated protein kinase; MMP, matrix metalloproteinase; PLCH,
pulmonary Langerhans cell histiocytosis.
cheobronchial epithelium) and are characterized by the pres-
ence of cell surface markers CD1a and CD207 (langerin).2 ligand, expressed by airway epithelial cells.21 As DCs mature,
The earliest lesion in PLCH involves the accumulation and CCR6 expression diminishes and CCR7 expression predom-
invasion of distal bronchiolar walls by abnormal CD1aþ DCs. inates.22 Mature DCs then exit the lung and migrate to the
The exact factors that drive the recruitment and subsequent draining lymph nodes along a CCR7 ligand-driven chemokine
accumulation of the DCs in PLCH are not fully characterized. gradient of CCL19 and CCL21.23 It has been demonstrated
Immature DCs express CCR6 and migrate toward the CCL20 that cigarette smoke affects DC numbers and maturity by

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


Pulmonary Langerhans Cell Histiocytosis Shaw et al. 271

increasing the expression of several proinflammatory medi- Smoking induces accumulation of CD1aþ cells in patients
ators in the lung.24–26 However, the effects of cigarette smoke with healthy lungs and patients with COPD.36–38 Production of
on DC function and pulmonary accumulation in PLCH are the glycoprotein, osteopontin, is increased with exposure to
unknown.27 Several chemokines including CCL5, CCL20, and nicotine, and it is found in high levels in bronchoalveolar lavage
CXCL11 have been identified as candidates with the potential (BAL) fluid in smokers with PLCH and may act as a chemo-
to drive DC accumulation in nonpulmonary LCH lesions, but attractant to monocytes, macrophages, and DCs.14 Cigarette
these have not been confirmed in PLCH.28 The lesions in PLCH smoke also stimulates local production of cytokines (TNF-α,
are focal, which suggests that changes in the bronchiolar GM-CSF, transforming growth factor-β, and CCL20) that are
epithelial microenvironment are essential to the accumula- important for the recruitment and differentiation of DCs, and
tion of the DCs.4 Evaluation of the loosely formed DC have been reported to be elevated in PLCH lesions.39–42
aggregates around the small airways has revealed that there However, the fact that only a minority of individuals who
are a portion of cells that produce CD1a, but not CD207 smoke cigarettes go on to develop PLCH suggests that cigarette
(langerin), indicating that the cells are undergoing matura- smoke is unlikely to be the sole causative agent for PLCH, and
tion and differentiation.14 rather acts as a potentiator in the pathogenic pathway in
genetically susceptible individuals.
Secondary Immune Activation
The CD1aþ DCs in PLCH express multiple costimulatory PLCH Is an Inflammatory Neoplasm
molecules, and by producing cytokines such as interleukin There has been significant debate over whether PLCH is a

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


1-β, granulocyte macrophage colony-stimulating factor neoplastic disorder or an inflammatory disease. Previously, it
(GM-CSF), and tumor necrosis factor (TNF)-α, they help was thought that PLCH is caused by cigarette smoke-induced
in secondary recruitment of other immune cells, forming polyclonal expansion of the DCs.43 However, recent data
cellular nodules comprised DCs, T-cells, B-cells, fibroblasts, have revealed the DCs in PLCH to be of clonal origin,44–46
eosinophils, neutrophils, plasma cells, and pigmented mac- thus suggesting a neoplastic nature for PLCH. Finally, the
rophages.18,29,30 As the disease progresses, the nodules are discovery of mutations involving the MAPK pathway in PLCH
gradually replaced by fibroblastic proliferation with central has transformed our understanding of the field and firmly
scarring and peripheral cellular tentacles.31 established PLCH as a neoplasm. The MAPK mutations found
in the PLCH lesions are similar to mutations found in other
Lung Destruction in PLCH malignancies such as melanoma,47 hairy cell leukemia,48
A unique characteristic of PLCH nodules is their ability to papillary thyroid cancer,49 as well as other histiocytic dis-
replace and remodel the surrounding tissues. The exact mech- orders such as ECD.50 The most common mutation described
anisms responsible for lung destruction in PLCH are not well in PLCH is the BRAFV600E mutation, present in 50% of the
understood. CD1aþ cells of PLCH nodules express membrane patients.51,52 Mourah et al analyzed biopsies in adult LCH
maturation markers that are similar to ones that are on the patients (26 pulmonary lesions and 37 nonpulmonary
surface of DCs after being activated by cytokines or patho- lesions) and found some key differences in the genetic
gens.32 One of the proposed mechanisms for the destruction landscape of PLCH as compared with the nonpulmonary
and remodeling is direct, local cytotoxic damage by pulmonary lesions of LCH.51 Similar to systemic LCH, mutations involv-
macrophage, natural killer cells, and cytotoxic CD8 T-cells that ing BRAFV600E and MAP2K1 were seen in 50 and 20% of the
have been aberrantly activated by the abnormal DCs. This is pulmonary lesions, respectively. In addition, the pulmonary
demonstrated by immunohistochemical analyses of PLCH lesions were found to have a separate DC clone harboring the
cellular lesions.20 Recent research has discovered several NRASQ61K/R mutation, often concurrently along with a
different metalloproteinases in LCH nodules, and it is BRAFV600E mutation. NRAS mutations were not identified in
theorized that this could contribute to, or be the primary the nonpulmonary lesions.
underlying cause of, the tissue destruction.14,33,34 Progressive The exact molecular mechanisms by which MAPK muta-
centrilobular destruction with fibrosis leads to the formation tions drive abnormal DC accumulation in PLCH is not fully
of the hallmark cystic airspace dilation that is characteristic of understood and represents a high priority area of future
PLCH. research. The CD1aþ cells do not appear to proliferate
abnormally53,54; instead these cells exhibit increased sur-
Role of Cigarette Smoke vival and are resistant to apoptosis.55 Other potential patho-
There is a very high prevalence of smoking in patients with genic mechanisms proposed in PLCH include activated MAPK
PLCH, which suggests a causal relationship.1,4,5,15 The vast pathway-induced senescence of the myeloid DCs,56 and
majority of the patients (90–100%) diagnosed with PLCH reduced migration of DCs from lungs to the lymph nodes.
are current or former smokers.4,16 A significant proportion of In summary, the current understanding of PLCH can be
patients who were diagnosed with LCH in childhood and conceptualized as a state of universal ERK activation due to
later developed PLCH had a history of cigarette smoking as upstream mutations involving one or more oncogenes in the
opposed to the patients who only had LCH and did not MAPK pathway. We propose that PLCH is a myeloid neoplasm
develop lung involvement.35 Cigarette smoke likely serves that follows the classic two-hit paradigm of malignancy as
as the inciting external antigen in patients with PLCH that described by Knudson,57 where genetically susceptible indi-
leads to an uncontrolled response by the DCs. viduals harboring MAPK mutations in the DCs develop PLCH

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


272 Pulmonary Langerhans Cell Histiocytosis Shaw et al.

after being exposed to an external environmental stimulus Imaging


(i.e., the second hit, cigarette/marijuana smoke).
PLCH lesions are classically bilateral, symmetric, and charac-
terized by upper lobe predominant nodules and cysts which
Clinical Features
spare the lung bases near the costophrenic sulci
Patients with PLCH can have wide-ranging presentations, (►Fig. 2).31,65,66 PLCH lesions are focal, located both centrally
from asymptomatic disease detected incidentally to pro- and peripherally, and are separated by normal-appearing
gressive debilitating disease. Initial symptoms can be insid- parenchyma.18 In the early stages of disease, imaging may
ious onset of dyspnea on exertion or cough, and it is not show bilateral, upper lobe predominant irregular, small
uncommon for patients to attribute these symptoms to nodules ranging between 1 and 10 mm in size.5 As disease
smoking.4,16 Approximately two-thirds of patients present progresses, the nodules start to develop cystic changes and
with dry cough and dyspnea on exertion as the initial both thick-walled cavities and thin-walled cysts can be
complaint.3,11 In a recent survey-based assessment of visualized on chest computed tomography (CT) scans. The
patients with PLCH, dyspnea on exertion was the most cysts in PLCH will initially appear to be round, small, and
common symptom (described in 89% of the patients), have thick walls; however, as the disease progresses, the
followed by fatigue (74%) and dry cough (72%).58 About cysts will start to have thinner walls and become larger and
10 to 20% of patients can present with constitutional irregular, bilobed, cloverleafed, or branched.31 The irregular-
symptoms such as malaise, fever, night sweats, and weight ity of the cysts as the disease progresses is thought to be due

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


loss.18 About 25% of PLCH patients are asymptomatic to the coalescence of the nodules.16 Mediastinal lymphade-
and are diagnosed based on incidental imaging nopathy is not a common finding in PLCH and should prompt
findings.8,9,12,59,60 Spontaneous pneumothorax, either uni- workup for an additional/alternate diagnosis.31 In later
lateral or bilateral, can be the presenting manifestation in stages, the disease can get “burnt out” and evolve into an
10 to 20% of patients.5,60,61 Pulmonary hypertension in upper lung predominant reticular pattern with fibrosis, and
patients with PLCH tends to be worse than pulmonary can be difficult to differentiate from other fibrosing intersti-
hypertension observed in comparable advanced diffuse tial lung diseases.31 Longitudinal studies involving serial
lung diseases.62,63 This is likely due to the direct involve- high-resolution CT (HRCT) scans have shown that over
ment of the small pulmonary arteries and arterioles by the time the nodules and cavitated nodules can resolve, but
disease process, rather than just loss of a portion of the the cysts tend to persist or enlarge over time.67 Ground-
cross-sectional area of the pulmonary circulation that is glass opacities, linear densities, and emphysematous bullae
seen with other diffuse lung diseases. Although hemosider- can also be seen, and reflect the concomitant presence of
in-laden macrophages may be seen histologically, hemop- cigarette smoke-induced lung damage in addition to PLCH.68
tysis is an uncommon symptom and should not be In pediatric patients with lung involvement from systemic
attributed to PLCH unless other etiologies, specifically LCH, it is not uncommon to have lesions present diffusely,
malignancies, have been ruled out.4,7,8,11,61,64 including the lung bases.69

Fig. 2 Imaging findings in PLCH: (A) Coronal CT chest demonstrating nodules, thick-walled cavities, and thin-walled cysts in an upper-mid lung
zone predominance with relative sparing of the lung bases. (B) Repeat CT chest performed 7 months following reduction in cigarette smoke
exposure demonstrating significant radiological improvement with almost complete resolution of the nodules and thick-walled cavities and
persistent cysts. CT, computed tomography; PLCH, pulmonary Langerhans cell histiocytosis.

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


Pulmonary Langerhans Cell Histiocytosis Shaw et al. 273

Pulmonary Function Testing presence of a convoluted nuclear membrane, and a nuclear


groove may also be visualized in some of the LCs. The nucleus
In early disease, pulmonary function tests (PFTs) can be normal lacks a prominent nucleolus and is surrounded by abundant
in 10% of patients with PLCH.70 Patients can have restrictive, cytoplasm, with poorly defined cellular borders.71,72 Diagnosis
obstructive, or combined defects on PFTs; the most common is confirmed by immunohistochemical staining for CD1a,
defect is decreased diffusing capacity of lung for carbon CD207 (langerin), and S-100.15 Birbeck granules on electron
monoxide (DLCO), seen in 70 to 90% of patients.3,5–7,11,12 microscope are characteristic of LCs and helpful for diagnosis
Almost all patients have an obstructive pattern on PFTs that is of PLCH; however, this mode of confirmation is not commonly
out of proportion to the smoking history, likely a reflection of utilized in clinical practice due to the associated expense and
small airway damage.3,5,7,11 A purely restrictive pattern is the fact that immunohistochemical staining for cell surface
uncommon but can occur.3 markers such as CD1a is readily available.16,71

Pathology Diagnosis
On gross examination, early lesions in PLCH are focal, tan- Definitive diagnosis of PLCH requires histopathological confir-
white nodules, and have irregular borders.71 On microscopic mation, either from the lungs (most common) or other extrap-
examination, lesions are centered around terminal and respi- ulmonary locations, typically skin or bone.4 In some cases,
ratory bronchioles and can be seen infiltrating the bronchiolar however, a combination of characteristic HRCT findings in the

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


walls and epithelium.72 As PLCH lesions grow, they progres- presence of a compatible clinical history, especially in patients
sively envelope the terminal bronchioles.73 The increasing with a history of exposure to cigarette smoke, may be sufficient
obstruction of the small airways that results from this process to establish a clinical diagnosis of PLCH.
is thought to be responsible for the distal perilesional airspace
enlargement and cyst formation.74 As the disease progresses, Bronchoalveolar Lavage
the lesions evolve into stellate fibrotic scars that are sur- The presence of more than 5% CD1aþ cells on BAL has been
rounded by cystic areas and honeycombing (►Fig. 3).71,72 proposed as a confirmatory diagnostic finding in patients
Histologically, the hallmark of PLCH is the accumulation of with suspected PLCH. However, this cutoff, while being quite
CD1aþ cells organized into aggregates in the nodules and specific, suffers from a lack of sensitivity and is not clinically
airspaces comprising the PLCH lesions (►Fig. 3). The CD1aþ useful in most patients. As such, BAL has limited utility in
cells are accompanied by variable numbers of eosinophils, establishing the diagnosis of PLCH but can be of value in
macrophages, plasma cells, and lymphocytes.52,73 A key his- some patients for excluding other etiologies of upper lobe
tological feature of LCs, which can help in identification, is the predominant nodules and cysts, such as atypical infections.

Fig. 3 Histopathological findings in PLCH: (A) Stellate inflammatory and fibrotic nodule with peripheral accumulation of smokers’ macrophages.
(B) Older lesion consisting of fibrotically thickened interstitial “tentacles” with accompanying inflammation and cystic airspace enlargement at
the periphery on the right. (C) Inflammatory infiltrate from the center of an active nodule, including Langerhans cells, conspicuous eosinophils,
and other inflammatory cells. (D) Langerhans cells (arrowhead) with ovoid cell shapes and folded or indented nuclei, some of which have
grooves. (E) High-power image of (B) highlighting the fibrotic and inflamed interstitium, and Langerhans cells and macrophages in an alveolus.
(F) Numerous Langerhans cells demonstrate brown membranous staining for CD1a. PLCH, pulmonary Langerhans cell histiocytosis.

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


274 Pulmonary Langerhans Cell Histiocytosis Shaw et al.

Lung Biopsy patients with severe/progressive disease. Inhaled cortico-


Transbronchial forceps-guided lung biopsy can establish the steroids in addition to bronchodilators can be tried, espe-
diagnosis of PLCH in approximately one-third of the cases. cially in patients with reversible defects or obstruction on
False negatives occur due to inadequate sampling of lesions or PFTs.1,4
interpretative challenges. A critical review of the biopsy speci- Vinblastine, and sometimes methotrexate, has been used
mens by an expert pathologist familiar with the disease in severe multisystemic LCH with some efficacy; however,
manifestations is essential prior to determining the procedure these drugs have not been shown to have a consistently
to be nondiagnostic. Transbronchial cryobiopsy of the lung is a positive effect in patients with isolated PLCH.4,78 Cladribine
relatively new procedure where the lung parenchyma is frozen (2-chlorodeoxyadenosine) has been shown to be helpful in
by using cold compressed gas and taken out en bloc. This refractory systemic LCH and has also been used in isolated
procedure yields bigger tissue specimens than the traditional PLCH.79–81 It is directly toxic to monocytes and lymphocytes
forceps biopsies and has been shown to have a diagnostic yield and can induce remission or improve pulmonary function in
approaching 70% in patients with diffuse parenchymal lung PLCH.81–87 A phase II clinical trial investigating the safety and
diseases. The diagnostic utility of transbronchial cryobiopsy in efficacy of cladribine in PLCH is currently underway
patents with PLCH is not well established; however, it is quite (NCT01473797, www.ClinicalTrials.gov).
likely to be clinically useful in patients with suspected PLCH The recent discovery of multiple MAPK mutations in PLCH
given the bronchiolocentric nature of the disease. Expert has suggested the possibility of targeted therapeutic inter-
centers familiar with this technique should consider trans- ventions similar to other histiocytic disorders and malignan-

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


bronchial lung cryobiopsy in patients with suspected PLCH cies harboring activating mutations in the MAPK pathway.
prior to performing surgical lung biopsy. Vemurafenib, a BRAF kinase inhibitor, has shown clinical
The gold standard diagnostic modality in patients with efficacy in patients with ECD and was recently approved by
suspected PLCH remains video-assisted thoracoscopic sur- the U.S. Food and Drug Administration to treat ECD.88 Given
gery–guided surgical lung biopsy and may be needed in some the similarities in pathogenesis, especially considering the
instances where all other non- or less-invasive forms of central role of BRAF mutations in both PLCH and ECD,
diagnostic confirmation have been exhausted. In a subset treatment with BRAF inhibition may be a clinically useful
of patients who present with a spontaneous pneumothorax, future strategy in PLCH. A combination of BRAF and MEK
the biopsy may be done at the same time as pleurodesis. inhibition has shown promising results in patients with
malignant melanoma, and may represent a viable treatment
approach in patients harboring concurrent BRAF and NRAS
Management
mutations.47,51 Excellent response using an oral MEK inhibi-
Smoking Cessation tor, cobimetinib, has recently been described in a diverse
Given the central role of cigarette smoke exposure in disease category of histiocytic disorders.89 The use of a MEK inhibitor
pathogenesis, smoking cessation is an essential component (trametinib) has also been described in a patient with
of management in patients with PLCH. In a substantial progressive PLCH harboring a MAP2K1 mutation, with
proportion of patients, smoking cessation alone may lead significant improvement in dyspnea and 6-minute walk
to disease regression and/or stabilization without the need distance.90 All of these studies are building on a small, but
for any other pharmacological treatment.1,61,75–77 The growing body of evidence that targeted therapies directed at
patients should be closely monitored, though, as smoking MAPK mutations could have a significant impact on the
cessation may be short lived and in a subset of patients treatment paradigm of PLCH in the near future.
(approximately one-third), the disease may progress despite
successful smoking cessation. It is important to note that Spontaneous Pneumothorax
while discussing smoking cessation, clinicians take into Spontaneous pneumothorax is a common pulmonary compli-
account both active and passive smoke exposure, as well as cation of PLCH, with reported prevalence estimates ranging
marijuana smoking. between 16 and 32%.58,61,91 In a subset of patients, spontane-
ous pneumothorax can be the presenting manifestation of
Pharmacological Treatment PLCH, and it is important to maintain a high index of clinical
Systemic corticosteroids are often prescribed to patients suspicion for underlying secondary causes when evaluating
with PLCH. Although there have been reports of symptomatic patients with an apparent primary spontaneous pneumotho-
and radiological improvement after systemic corticosteroids, rax. Patients with PLCH should be educated about the signs and
these reports suffer from a less-than-perfect characteriza- symptoms of a spontaneous pneumothorax and advised to seek
tion of the timing of corticosteroid initiation with respect to immediate medical attention in the event that they experience
smoking cessation, and it is possible that these patients may these symptoms. Spontaneous pneumothorax in PLCH tends to
have improved by smoking cessation alone rather than a true recur frequently, if managed conservatively, with reported
treatment effect of corticosteroids. Long-term administra- recurrence rates approaching 60%.58,61 Surgical pleurodesis
tion of corticosteroids is associated with a plethora of can substantially reduce the risk of recurrence to 0 to 20%58,61
adverse effects. Given the overall lack of evidence, we typi- and should be considered following the first episode of
cally do not prescribe systemic corticosteroids routinely for spontaneous pneumothorax rather than waiting for a recurrent
patients with PLCH, except for a short, well-defined trial in episode. The risk of a spontaneous pneumothorax associated

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


Pulmonary Langerhans Cell Histiocytosis Shaw et al. 275

with air travel is less than 1 per 100 flights, and air travel is who underwent lung transplantation in the United States
considered safe for majority of the patients with PLCH. over the past three decades. The median posttransplant
survival in patients with PLCH was similar to other lung
General Recommendations diseases (5.1 years in the PLCH group vs. 5.5 years in patients
Overall, smoking cessation remains the focal point of man- with other lung diseases), with actuarial Kaplan–Meier 1-, 5-,
agement for most patients with PLCH. Attention should be and 10-year survival of 85, 49, and 22%, respectively. For
paid toward optimal vaccinations including annual influenza reasons that are not entirely clear, females had a significantly
vaccination and appropriate pneumococcal vaccination increased median survival as compared with males (9.3 vs.
using both PPSV23 and PCV13. Some patients with PLCH 3.9 years, p ¼ 87).100 Similar to other lung transplantation
will require supplemental oxygen therapy; recommenda- recipients, bronchiolitis obliterans syndrome was the lead-
tions for prescribing supplemental oxygen to patients with ing cause of posttransplant morbidity and mortality in
PLCH are the same as other chronic lung diseases. Pulmonary patients with PLCH.100
hypertension in PLCH portends a worse prognosis, and
patients should be monitored periodically for signs and Monitoring Disease Extent and Progression
symptoms suggestive of pulmonary hypertension. History The natural history of PLCH can be quite variable in individual
taking in PLCH should include enquiries about extrapulmo- patients. All patients with newly diagnosed PLCH should have
nary organ involvement such as weight loss, polyuria, poly- close follow-up with serial PFTs every 3 to 4 months at least for
dipsia, bone pain, and skin rashes. There are some reports of the first 1 to 2 years, to confidently establish their individual

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


worsening diabetes insipidus in pregnant females; however, disease trajectory. Subsequently, the follow-up PFTs can be
PLCH is not a contraindication to pregnancy unless associat- spaced to every 6 months, and then perhaps annually after
ed with pulmonary hypertension and/or chronic respiratory establishing a stable trajectory.70,101,102 Rapid decline in
failure.92,93 Key recommendations for management of forced expiratory volume in 1 second (FEV1) and/or changes
patients with PLCH are summarized in ►Table 1. in functional status should prompt consideration of obtaining
a repeat CTof the chest to evaluate disease progression. History
Lung Transplantation and physical examination aimed at ascertaining extrapulmo-
Lung transplantation has been used to treat patients with nary organ involvement from PLCH (diabetes insipidus, skin
end-stage PLCH and patients with severe pulmonary hyper- rashes, and bone pain) or other disease-specific complications
tension with some success. Disease recurrence following such as pulmonary hypertension should be performed at the
lung transplantation has been described in several cases, initial and each subsequent encounter. Laboratory tests that
highlighting the neoplastic nature of PLCH where the abnor- might be useful in patients with PLCH include tests aimed at
mal DCs carrying the MAPK mutations originate from their monitoring renal function and electrolyte imbalances, as well
hematopoietic precursors and metastasize to the lungs.94–98 as involvement of high-risk organs (spleen, liver, and bone
In 2006, Dauriat et al reviewed data from 39 PLCH patients marrow) (►Table 2).
who had been transplanted in France, and found the 1-, 5-,
and 10-year posttransplant survival rates to be 77, 57, and
Natural History and Prognosis
54%, respectively.99 Recurrent disease was seen in 20% of
the transplanted patients.99 Recently, Wajda et al analyzed Disease course in PLCH is highly variable and unpredictable.
the posttransplantation outcomes from 87 PLCH patients Published reports suggest a median survival of 12 to 13 years

Table 1 Recommendations for optimal management of patients with PLCH

Obtain detailed history with regard to smoke exposure, cigarettes (both active and passive), as well as marijuana, and counsel
patients on complete cessation of active smoking and avoidance of passive smoking
Enquire about extrapulmonary symptoms at each visit: weight loss, polyuria, polydipsia, bone pain, and skin rashes
Pay attention to the signs and symptoms of pulmonary hypertension
Make attempts to ascertain the underlying genetic mutation (in patients with available tissue specimens)
Consider a trial of bronchodilators and inhaled corticosteroids in symptomatic patients with obstructive defect on PFTs
Consider pharmacological treatment with cladribine or targeted treatment with BRAF (in patients with known single BRAFV600E
mutation) or MEK inhibitors (in patients with other MAPK mutations or unknown mutation status). Candidates for pharma-
cological treatment should be chosen after a thorough and careful consideration of the risk-to-benefit ratio of treatment. For
instance, potential candidates may include patients with progressive disease despite successful smoking cessation, or patients
with multisystem LCH especially with risk-organ involvement
Educate patients about the signs and symptoms of a spontaneous pneumothorax
Educate patients about the safety of air travel and counsel to avoid scuba diving
Annual influenza vaccinations and appropriate pneumococcal vaccinations (both PCV13 and PPSV23)

Abbreviations: LCH, Langerhans cell histiocytosis; MAPK, mitogen-activated protein kinase; PCV13, pneumococcal conjugate vaccine; PFT,
pulmonary function test; PLCH, pulmonary Langerhans cell histiocytosis; PPSV23, pneumococcal polysaccharide vaccine.

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


276 Pulmonary Langerhans Cell Histiocytosis Shaw et al.

Table 2 Disease monitoring in patients with PLCH

Initial visit Every 3–4 mo until Annual visit Focused assessments based
clinical stability following stability on symptoms and exam findings
Enquire about symptoms of X X X
extrapulmonary involvement
PFTs X X X
CBC, renal and liver X X
function tests
CT chest X X
Echocardiogram X
Brain MRI X
PET scan X
Bone scan X

Abbreviations: CBC, complete blood count; CT, computed tomography; MRI, magnetic resonance imaging; PET, positron emission tomography; PFT,
pulmonary function test; PLCH, pulmonary Langerhans cell histiocytosis.

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


following disease diagnosis.3,14 However, the mortality and optimal dosing strategies as well as determine their safety
morbidity in PLCH are likely overestimated in the literature, and efficacy in this patient population.
as a significant proportion of patients with mild disease are
lost to follow-up.3,4,18,64 About 50% of patients with PLCH
Conclusion
will experience a favorable outcome.103 About 10 to 20% of
patients have early severe symptoms that consist of recur- PLCH is a cigarette smoke-driven inflammatory myeloid neo-
rent pneumothoraces or progressive respiratory failure with plasm, where DCs harboring pathogenic MAPK mutations
cor pulmonale.4,18 Approximately 30 to 40% of patients have drive progressive nodulocystic lung destruction. Improved
persistent symptoms of variable severity with conversion of understanding of the underlying genetic landscape of PLCH
radiographic findings from nodules to cysts over time.4 has transformed and energized the field, and provided a
Predictors of poor outcomes are diagnosis at older age, promise for accelerated scientific discoveries, disease-specific
multisystem involvement (especially involvement of the biomarkers, targeted therapies, and overall improved patient
liver, spleen, and/or bone marrow), extensive cysts and outcomes.
honeycombing on imaging, and abnormal PFT parameters
such as reduced DLCO, low FEV1/forced vital capacity ratio at Conflict of Interest
diagnosis, and high residual volume/total lung capacity ratio None declared.
at diagnosis.3,61 In a prospective, multicenter natural history
study of 58 patients with newly diagnosed PLCH, ongoing
References
cigarette smoking and reduced PaO2 were associated with a
1 Wei P, Lu HW, Jiang S, Fan LC, Li HP, Xu JF. Pulmonary Langerhans
faster rate of lung function decline.70 cell histiocytosis: case series and literature review. Medicine
(Baltimore) 2014;93(23):e141
2 Emile JF, Abla O, Fraitag S, et al; Histiocyte Society. Revised
Future Needs classification of histiocytoses and neoplasms of the macro-
There are several gaps in our understanding of the disease phage-dendritic cell lineages. Blood 2016;127(22):2672–2681
3 Delobbe A, Durieu J, Duhamel A, Wallaert B. Determinants of
biology that have hindered meaningful progress in PLCH.
survival in pulmonary Langerhans’ cell granulomatosis (histiocy-
Perhaps, the most important need for the field is an improved tosis X). Groupe d’Etude en Pathologie Interstitielle de la Société de
understanding of the disease pathogenesis. Improved under- Pathologie Thoracique du Nord. Eur Respir J 1996;9(10):2002–2006
standing of the role of MAPK mutations and their effect on DC 4 Tazi A. Adult pulmonary Langerhans’ cell histiocytosis. Eur
function, and the exact role of cigarette smoke exposure on Respir J 2006;27(06):1272–1285
5 Vassallo R, Ryu JH, Schroeder DR, Decker PA, Limper AH. Clinical
the disease pathogenesis will help develop novel therapies.
outcomes of pulmonary Langerhans’-cell histiocytosis in adults.
The development of disease-specific diagnostic biomarkers N Engl J Med 2002;346(07):484–490
can obviate the need for invasive procedures such as lung 6 Basset F, Corrin B, Spencer H, et al. Pulmonary histiocytosis X. Am
biopsies to establish the diagnosis with certainty. Prospec- Rev Respir Dis 1978;118(05):811–820
tive, natural history studies are required to determine pre- 7 Crausman RS, Jennings CA, Tuder RM, Ackerson LM, Irvin CG,
dictors of disease progression that can help design pivotal King TE Jr. Pulmonary histiocytosis X: pulmonary function and
exercise pathophysiology. Am J Respir Crit Care Med 1996;153
treatment trials evaluating novel agents and determine the
(01):426–435
optimal population to be included in these trials. Finally, 8 Friedman PJ, Liebow AA, Sokoloff J. Eosinophilic granuloma of
targeted therapies aimed at MAPK pathway inhibition need lung. Clinical aspects of primary histiocytosis in the adult.
to be investigated in a controlled fashion to understand Medicine (Baltimore) 1981;60(06):385–396

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


Pulmonary Langerhans Cell Histiocytosis Shaw et al. 277

9 Howarth DM, Gilchrist GS, Mullan BP, Wiseman GA, Edmonson 29 Castoldi MC, Verrioli A, De Juli E, Vanzulli A. Pulmonary Langer-
JH, Schomberg PJ. Langerhans cell histiocytosis: diagnosis, natu- hans cell histiocytosis: the many faces of presentation at initial
ral history, management, and outcome. Cancer 1999;85(10): CT scan. Insights Imaging 2014;5(04):483–492
2278–2290 30 Leatherwood DL, Heitkamp DE, Emerson RE. Best cases from the
10 Schönfeld N, Frank W, Wenig S, et al. Clinical and radiologic AFIP: pulmonary Langerhans cell histiocytosis. Radiographics
features, lung function and therapeutic results in pulmonary 2007;27(01):265–268
histiocytosis X. Respiration 1993;60(01):38–44 31 Stone S, Lynch D. National Jewish Health classical pulmonary
11 Travis WD, Borok Z, Roum JH, et al. Pulmonary Langerhans cell radiology case reports: imaging features of Langerhans cell
granulomatosis (histiocytosis X). A clinicopathologic study of 48 histiocytosis. QJM 2018;111(10):739–740
cases. Am J Surg Pathol 1993;17(10):971–986 32 Geissmann F, Lepelletier Y, Fraitag S, et al. Differentiation of
12 Watanabe R, Tatsumi K, Hashimoto S, Tamakoshi A, Kuriyama T; Langerhans cells in Langerhans cell histiocytosis. Blood 2001;97
Respiratory Failure Research Group of Japan. Clinico-epidemio- (05):1241–1248
logical features of pulmonary histiocytosis X. Intern Med 2001; 33 de Graaf JH, Tamminga RY, Dam-Meiring A, Kamps WA, Timens
40(10):998–1003 W. The presence of cytokines in Langerhans’ cell histiocytosis.
13 Haupt R, Minkov M, Astigarraga I, et al; Euro Histio Network. J Pathol 1996;180(04):400–406
Langerhans cell histiocytosis (LCH): guidelines for diagnosis, 34 Hayashi T, Rush WL, Travis WD, Liotta LA, Stetler-Stevenson WG,
clinical work-up, and treatment for patients till the age of 18 Ferrans VJ. Immunohistochemical study of matrix metallopro-
years. Pediatr Blood Cancer 2013;60(02):175–184 teinases and their tissue inhibitors in pulmonary Langerhans’
14 Vassallo R, Harari S, Tazi A. Current understanding and manage- cell granulomatosis. Arch Pathol Lab Med 1997;121(09):
ment of pulmonary Langerhans cell histiocytosis. Thorax 2017; 930–937
72(10):937–945 35 Bernstrand C, Cederlund K, Sandstedt B, et al. Pulmonary abnor-

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


15 Kim T, Kwon HJ, Eom M, Kim SW, Sin MH, Jung SH. Bronchial malities at long-term follow-up of patients with Langerhans cell
washing cytology of pulmonary Langerhans cell histiocytosis: a histiocytosis. Med Pediatr Oncol 2001;36(04):459–468
case report. J Pathol Transl Med 2017;51(04):444–447 36 Hashimoto M, Yanagisawa H, Minagawa S, et al. TGF-β-depen-
16 Juvet SC, Hwang D, Downey GP. Rare lung diseases III: pulmonary dent dendritic cell chemokinesis in murine models of airway
Langerhans’ cell histiocytosis. Can Respir J 2010;17(03):e55–e62 disease. J Immunol 2015;195(03):1182–1190
17 Gaensler EA, Carrington CB. Open biopsy for chronic diffuse 37 Soler P, Moreau A, Basset F, Hance AJ. Cigarette smoking-induced
infiltrative lung disease: clinical, roentgenographic, and physio- changes in the number and differentiated state of pulmonary
logical correlations in 502 patients. Ann Thorac Surg 1980;30 dendritic cells/Langerhans cells. Am Rev Respir Dis 1989;139
(05):411–426 (05):1112–1117
18 Lorillon G, Tazi A. How I manage pulmonary Langerhans cell 38 Vassallo R, Walters PR, Lamont J, Kottom TJ, Yi ES, Limper AH.
histiocytosis. Eur Respir Rev 2017;26(145):170070 Cigarette smoke promotes dendritic cell accumulation in COPD;
19 Thomeer M, Demedts M, Vandeurzen K; VRGT Working Group on a Lung Tissue Research Consortium study. Respir Res 2010;11:45
Interstitial Lung Diseases. Registration of interstitial lung dis- 39 Bracke KR, D’hulst AI, Maes T, et al. Cigarette smoke-induced
eases by 20 centres of respiratory medicine in Flanders. Acta Clin pulmonary inflammation and emphysema are attenuated in
Belg 2001;56(03):163–172 CCR6-deficient mice. J Immunol 2006;177(07):4350–4359
20 Nagarjun Rao R, Chang CC, Tomashefski JF Jr. Lymphocyte sub- 40 Marchal J, Kambouchner M, Tazi A, Valeyre D, Soler P. Expression
populations and non-Langerhans’ cell monocytoid cells in pul- of apoptosis-regulatory proteins in lesions of pulmonary Lan-
monary Langerhans’ cell histiocytosis. Pathol Res Pract 2008;204 gerhans cell histiocytosis. Histopathology 2004;45(01):20–28
(05):315–322 41 Prasse A, Stahl M, Schulz G, et al. Essential role of osteopontin in
21 Ito T, Carson WF IV, Cavassani KA, Connett JM, Kunkel SL. CCR6 as smoking-related interstitial lung diseases. Am J Pathol 2009;174
a mediator of immunity in the lung and gut. Exp Cell Res 2011; (05):1683–1691
317(05):613–619 42 Reibman J, Hsu Y, Chen LC, Bleck B, Gordon T. Airway epithelial
22 Sallusto F, Schaerli P, Loetscher P, et al. Rapid and coordinated cells release MIP-3alpha/CCL20 in response to cytokines and
switch in chemokine receptor expression during dendritic cell ambient particulate matter. Am J Respir Cell Mol Biol 2003;28
maturation. Eur J Immunol 1998;28(09):2760–2769 (06):648–654
23 Worbs T, Hammerschmidt SI, Förster R. Dendritic cell migration 43 Yousem SA, Colby TV, Chen YY, Chen WG, Weiss LM. Pulmonary
in health and disease. Nat Rev Immunol 2017;17(01):30–48 Langerhans’ cell histiocytosis: molecular analysis of clonality.
24 Botelho FM, Nikota JK, Bauer CM, et al. Cigarette smoke-induced Am J Surg Pathol 2001;25(05):630–636
accumulation of lung dendritic cells is interleukin-1α-depen- 44 Willman CL, Busque L, Griffith BB, et al. Langerhans’-cell histio-
dent in mice. Respir Res 2012;13:81 cytosis (histiocytosis X)–a clonal proliferative disease. N Engl J
25 Demedts IK, Bracke KR, Van Pottelberge G, et al. Accumulation of Med 1994;331(03):154–160
dendritic cells and increased CCL20 levels in the airways of 45 Willman CL, McClain KL. An update on clonality, cytokines, and
patients with chronic obstructive pulmonary disease. Am J viral etiology in Langerhans cell histiocytosis. Hematol Oncol
Respir Crit Care Med 2007;175(10):998–1005 Clin North Am 1998;12(02):407–416
26 Lommatzsch M, Bratke K, Knappe T, et al. Acute effects of tobacco 46 Yu RC, Chu C, Buluwela L, Chu AC. Clonal proliferation of
smoke on human airway dendritic cells in vivo. Eur Respir J 2010; Langerhans cells in Langerhans cell histiocytosis. Lancet 1994;
35(05):1130–1136 343(8900):767–768
27 Tsoumakidou M, Demedts IK, Brusselle GG, Jeffery PK. Dendritic 47 Eroglu Z, Ribas A. Combination therapy with BRAF and MEK
cells in chronic obstructive pulmonary disease: new players in inhibitors for melanoma: latest evidence and place in therapy.
an old game. Am J Respir Crit Care Med 2008;177(11): Ther Adv Med Oncol 2016;8(01):48–56
1180–1186 48 Durham BH, Getta B, Dietrich S, et al. Genomic analysis of hairy
28 Annels NE, Da Costa CE, Prins FA, Willemze A, Hogendoorn PC, cell leukemia identifies novel recurrent genetic alterations.
Egeler RM. Aberrant chemokine receptor expression and che- Blood 2017;130(14):1644–1648
mokine production by Langerhans cells underlies the pathogen- 49 Kim WW, Ha TK, Bae SK. Clinical implications of the BRAF
esis of Langerhans cell histiocytosis. J Exp Med 2003;197(10): mutation in papillary thyroid carcinoma and chronic lympho-
1385–1390 cytic thyroiditis. J Otolaryngol Head Neck Surg 2018;47(01):4

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


278 Pulmonary Langerhans Cell Histiocytosis Shaw et al.

50 Allen CE, Parsons DW. Biological and clinical significance of 73 Kambouchner M, Basset F, Marchal J, Uhl JF, Hance AJ, Soler P.
somatic mutations in Langerhans cell histiocytosis and related Three-dimensional characterization of pathologic lesions in
histiocytic neoplastic disorders. Hematology (Am Soc Hematol pulmonary Langerhans cell histiocytosis. Am J Respir Crit Care
Educ Program) 2015;2015:559–564 Med 2002;166(11):1483–1490
51 Mourah S, How-Kit A, Meignin V, et al. Recurrent NRAS muta- 74 Emile JF, Wechsler J, Brousse N, et al. Langerhans’ cell histiocy-
tions in pulmonary Langerhans cell histiocytosis. Eur Respir J tosis. Definitive diagnosis with the use of monoclonal antibody
2016;47(06):1785–1796 O10 on routinely paraffin-embedded samples. Am J Surg Pathol
52 Roden AC, Yi ES. Pulmonary Langerhans cell histiocytosis: an 1995;19(06):636–641
update from the pathologists’ perspective. Arch Pathol Lab Med 75 Mogulkoc N, Veral A, Bishop PW, Bayindir U, Pickering CA, Egan JJ.
2016;140(03):230–240 Pulmonary Langerhans’ cell histiocytosis: radiologic resolution
53 Brabencova E, Tazi A, Lorenzato M, et al. Langerhans cells in following smoking cessation. Chest 1999;115(05):1452–1455
Langerhans cell granulomatosis are not actively proliferating 76 Tazi A, Montcelly L, Bergeron A, Valeyre D, Battesti JP, Hance AJ.
cells. Am J Pathol 1998;152(05):1143–1149 Relapsing nodular lesions in the course of adult pulmonary
54 Senechal B, Elain G, Jeziorski E, et al. Expansion of regulatory T Langerhans cell histiocytosis. Am J Respir Crit Care Med 1998;
cells in patients with Langerhans cell histiocytosis. PLoS Med 157(6 Pt 1):2007–2010
2007;4(08):e253 77 Von Essen S, West W, Sitorius M, Rennard SI. Complete resolution
55 Caffrey PR, Altman RS. Pulmonary alveolar microlitbiasis occur- of roentgenographic changes in a patient with pulmonary
ring in premature twins. J Pediatr 1965;66:758–763 histiocytosis X. Chest 1990;98(03):765–767
56 Chilosi M, Facchetti F, Caliò A, et al. Oncogene-induced senes- 78 Tazi A, Lorillon G, Haroche J, et al. Vinblastine chemotherapy in
cence distinguishes indolent from aggressive forms of pulmo- adult patients with Langerhans cell histiocytosis: a multicenter
nary and non-pulmonary Langerhans cell histiocytosis. Leuk retrospective study. Orphanet J Rare Dis 2017;12(01):95

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.


Lymphoma 2014;55(11):2620–2626 79 Goh NS, McDonald CE, MacGregor DP, Pretto JJ, Brodie GN.
57 Knudson AG Jr. Mutation and cancer: statistical study of retino- Successful treatment of Langerhans cell histiocytosis with 2-
blastoma. Proc Natl Acad Sci U S A 1971;68(04):820–823 chlorodeoxyadenosine. Respirology 2003;8(01):91–94
58 Singla A, Kopras EJ, Gupta N. Spontaneous pneumothorax and air 80 Pardanani A, Phyliky RL, Li CY, Tefferi A. 2-Chlorodeoxyadeno-
travel in Pulmonary Langerhans cell histiocytosis: a patient sine therapy for disseminated Langerhans cell histiocytosis.
survey. Respir Investig 2019:S2212-5345(19)30148-0 Mayo Clin Proc 2003;78(03):301–306
59 Colby TV, Lombard C. Histiocytosis X in the lung. Hum Pathol 81 Saven A, Burian C. Cladribine activity in adult Langerhans-cell
1983;14(10):847–856 histiocytosis. Blood 1999;93(12):4125–4130
60 Minghini A, Trogdon SD. Recurrent spontaneous pneumothorax 82 Aerni MR, Aubry MC, Myers JL, Vassallo R. Complete remission of
in pulmonary histiocytosis X. Am Surg 1998;64(11):1040–1042 nodular pulmonary Langerhans cell histiocytosis lesions in-
61 Mendez JL, Nadrous HF, Vassallo R, Decker PA, Ryu JH. Pneumo- duced by 2-chlorodeoxyadenosine in a non-smoker. Respir
thorax in pulmonary Langerhans cell histiocytosis. Chest 2004; Med 2008;102(02):316–319
125(03):1028–1032 83 Epaud R, Ducou Le Pointe H, Fasola S, et al. Cladribine improves
62 Fartoukh M, Humbert M, Capron F, et al. Severe pulmonary lung cysts and pulmonary function in a child with histiocytosis.
hypertension in histiocytosis X. Am J Respir Crit Care Med 2000; Eur Respir J 2015;45(03):831–833
161(01):216–223 84 Grobost V, Khouatra C, Lazor R, Cordier JF, Cottin V. Effectiveness
63 Harari S, Brenot F, Barberis M, Simmoneau G. Advanced pulmo- of cladribine therapy in patients with pulmonary Langerhans cell
nary histiocytosis X is associated with severe pulmonary hyper- histiocytosis. Orphanet J Rare Dis 2014;9:191
tension. Chest 1997;111(04):1142–1144 85 Lazor R, Etienne-Mastroianni B, Khouatra C, Tazi A, Cottin V,
64 Vassallo R, Ryu JH, Colby TV, Hartman T, Limper AH. Pulmonary Cordier JF. Progressive diffuse pulmonary Langerhans cell his-
Langerhans’-cell histiocytosis. N Engl J Med 2000;342(26): tiocytosis improved by cladribine chemotherapy. Thorax 2009;
1969–1978 64(03):274–275
65 Abbott GF, Rosado-de-Christenson ML, Franks TJ, Frazier AA, 86 Lorillon G, Bergeron A, Detourmignies L, et al. Cladribine is
Galvin JR. From the archives of the AFIP: pulmonary Langerhans effective against cystic pulmonary Langerhans cell histiocytosis.
cell histiocytosis. Radiographics 2004;24(03):821–841 Am J Respir Crit Care Med 2012;186(09):930–932
66 Lacronique J, Roth C, Battesti JP, Basset F, Chretien J. Chest 87 Weitzman S, Braier J, Donadieu J, et al. 2′-Chlorodeoxyadenosine
radiological features of pulmonary histiocytosis X: a report (2-CdA) as salvage therapy for Langerhans cell histiocytosis
based on 50 adult cases. Thorax 1982;37(02):104–109 (LCH). Results of the LCH-S-98 protocol of the Histiocyte Society.
67 Brauner MW, Grenier P, Tijani K, Battesti JP, Valeyre D. Pulmo- Pediatr Blood Cancer 2009;53(07):1271–1276
nary Langerhans cell histiocytosis: evolution of lesions on CT 88 Diamond EL, Subbiah V, Lockhart AC, et al. Vemurafenib for BRAF
scans. Radiology 1997;204(02):497–502 V600-mutant Erdheim-Chester disease and Langerhans cell
68 Brauner MW, Grenier P, Mouelhi MM, Mompoint D, Lenoir S. histiocytosis: analysis of data from the histology-independent,
Pulmonary histiocytosis X: evaluation with high-resolution CT. phase 2, open-label VE-BASKET study. JAMA Oncol 2018;4(03):
Radiology 1989;172(01):255–258 384–388
69 Seely JM, Salahudeen S Sr, Cadaval-Goncalves AT, et al. Pulmo- 89 Diamond EL, Durham BH, Ulaner GA, et al. Efficacy of MEK
nary Langerhans cell histiocytosis: a comparative study of inhibition in patients with histiocytic neoplasms. Nature
computed tomography in children and adults. J Thorac Imaging 2019;567(7749):521–524
2012;27(01):65–70 90 Lorillon G, Jouenne F, Baroudjian B, et al. Response to trametinib
70 Tazi A, de Margerie C, Naccache JM, et al. The natural history of of a pulmonary Langerhans cell histiocytosis harboring a
adult pulmonary Langerhans cell histiocytosis: a prospective MAP2K1 deletion. Am J Respir Crit Care Med 2018;198(05):
multicentre study. Orphanet J Rare Dis 2015;10:30 675–678
71 Thurlbeck WM, Churg A. Thurlbeck’s Pathology of the Lung, 3rd 91 Radzikowska E, Błasińska-Przerwa K, Wiatr E, Bestry I, Langfort
ed. New York: Thieme; 2005 R, Roszkowski-Śliż K. Pneumothorax in patients with pulmonary
72 Lieberman PH, Jones CR, Steinman RM, et al. Langerhans cell Langerhans cell histiocytosis. Lung 2018;196(06):715–720
(eosinophilic) granulomatosis. A clinicopathologic study encom- 92 DiMaggio LA, Lippes HA, Lee RV. Histiocytosis X and pregnancy.
passing 50 years. Am J Surg Pathol 1996;20(05):519–552 Obstet Gynecol 1995;85(5 Pt 2):806–809

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020


Pulmonary Langerhans Cell Histiocytosis Shaw et al. 279

93 King TE Jr. Restrictive lung disease in pregnancy. Clin Chest Med 99 Dauriat G, Mal H, Thabut G, et al. Lung transplantation for
1992;13(04):607–622 pulmonary Langerhans’ cell histiocytosis: a multicenter analysis.
94 Boehler A. Lung transplantation for cystic lung diseases: lym- Transplantation 2006;81(05):746–750
phangioleiomyomatosis, histiocytosis x, and sarcoidosis. Semin 100 Wajda N, Zhu Z, Jandarov R, Dilling DF, Gupta N. Clinical
Respir Crit Care Med 2001;22(05):509–516 outcomes and survival following lung transplantation in
95 Etienne B, Bertocchi M, Gamondes JP, et al. Relapsing pulmonary patients with pulmonary Langerhans cell histiocytosis. Respir-
Langerhans cell histiocytosis after lung transplantation. Am J ology 2019
Respir Crit Care Med 1998;157(01):288–291 101 Girschikofsky M, Arico M, Castillo D, et al. Management of adult
96 Gabbay E, Dark JH, Ashcroft T, et al. Recurrence of Langerhans’ patients with Langerhans cell histiocytosis: recommendations
cell granulomatosis following lung transplantation. Thorax from an expert panel on behalf of Euro-Histio-Net. Orphanet J
1998;53(04):326–327 Rare Dis 2013;8:72
97 Habib SB, Congleton J, Carr D, et al. Recurrence of recipient 102 Suri HS, Yi ES, Nowakowski GS, Vassallo R. Pulmonary Langer-
Langerhans’ cell histiocytosis following bilateral lung transplan- hans cell histiocytosis. Orphanet J Rare Dis 2012;7:16
tation. Thorax 1998;53(04):323–325 103 Tazi A, Soler P, Hance AJ. Adult pulmonary Langerhans’ cell
98 Sulica R, Teirstein A, Padilla ML. Lung transplantation in inter- histiocytosis. Thorax 2000;55(05):405–416
stitial lung disease. Curr Opin Pulm Med 2001;7(05):314–322

Downloaded by: Universiteitsbibliotheek Amsterdam. Copyrighted material.

Seminars in Respiratory and Critical Care Medicine Vol. 41 No. 2/2020

You might also like