You are on page 1of 11

bs_bs_banner

Pediatrics International (2014) 56, 451–461 doi: 10.1111/ped.12380

Review Article

Recent advances in Langerhans cell histiocytosis

Akira Morimoto,1 Yukiko Oh,1 Yoko Shioda,2 Kazuko Kudo3 and Toshihiko Imamura4
1
Department of Pediatrics, Jichi Medical University of Medicine, Shimotsuke, 2Division of Hematology/Oncology, National
Center for Child Health and Development, Tokyo, 3Division of Hematology and Oncology, Shizuoka Children’s Hospital,
Shizuoka, and 4Department of Pediatrics, Kyoto Prefectural University of Medicine, Kyoto, Japan

Abstract The purpose of this review is to provide an updated overview of the pathogenesis and treatment of Langerhans cell
histiocytosis (LCH). The pathogenesis of LCH remains obscure and the optimal treatment for LCH has not been
established, although incremental progress has been made. Proinflammatory cytokines and chemokines are known to
play a role in LCH, which suggests that LCH is an immune disorder. However, the oncogenic BRAF mutation is also
detected in more than half of LCH patients, which suggests that LCH is a neoplastic disorder. Remaining major issues
in the treatment of LCH are how to rescue patients who have risk-organ involvement but do not respond to first-line
therapy, the optimal treatment for the orphan disease of multifocal adult LCH, and how to reduce and treat central
nervous system-related consequences, such as central diabetes insipidus and neurodegeneration. More research is needed
to better understand the pathogenesis of this disease and to resolve the treatment issues.

Key words BRAF, chemokine, cytokine, diabetes insipidus, Langerhans cell histiocytosis.

Langerhans cell histiocytosis (LCH), which was previously this age group between 0 and 3 years of age.1–3 It is estimated that
termed “histiocytosis X,” is a rare clonal disorder characterized 70 children are diagnosed with LCH newly on an annual basis in
by the proliferation of clonal CD1a-positive immature dendritic Japan.3 MS disease, especially RO-positive MS disease, is quite
cells (LCH cells) in the skin, bone, lymph nodes and other common in infants. SS disease accounts for about 70% of cases,
organs. The clinical manifestations of LCH vary from a self- while nearly 10% of cases have RO-positive MS disease. LCH
limiting single bone disease to rapidly fatal disseminated disease. appears to be more common in boys than in girls (1.2–2:1). In
However, LCH usually follows a chronic course and reactivations adults, the incidence is about one-third of the childhood inci-
often occur. This can result in permanent consequences, such as dence (i.e., 1–2 cases per million per year)4 and it occurs more
orthopedic abnormalities, the development of central diabetes frequently in young adults than in older adults.5 The development
insipidus (CDI), and neurodegenerative central nervous system of the disease is usually sporadic. However, genetic predisposi-
(ND-CNS) disease. LCH was originally described as three dif- tion may play a role in the development of LCH because about
ferent entities, namely, eosinophilic granuloma, Hand–Schüller– 1% of patients have relatives with LCH, monozygotic twin pairs
Christian disease, and Letterer–Siwe disease. However, at are concordant for LCH,6 and patients with LCH are more likely
present, LCH is classified as single-system (SS) vs multisystem to develop malignant disorders than the normal population.7
(MS) and unifocal vs multifocal disease. MS disease is classified
into two groups depending on whether risk organs (RO), namely, Pathology and diagnosis
the liver, lung, spleen and bone marrow, are involved. SS disease,
RO-negative MS disease, and RO-positive MS disease are almost To diagnose LCH, it is essential to perform a pathological exami-
equivalent to eosinophilic granuloma, Hand–Schüller–Christian nation. After hematoxylin–eosin staining, LCH cells have a dis-
disease, and Letterer–Siwe disease, respectively. This review tinctive homogeneously stained pink cytoplasm. The nuclei
largely describes the new insights into the pathogenesis and treat- appear twisted with a longitudinal groove and the small nucleolus
ment of LCH. has a “coffee bean” appearance. Immunohistochemical staining
of S-100 protein is helpful for detecting LCH cells. LCH cells do
Epidemiology express CD14, CD68 and intracellular major histocompatibility
complex (MHC) class II antigen, but do not express Fascin,
The incidence of LCH in children aged less than 15 years is 4–5
CD83 or CD86. A definitive diagnosis can be made when the
cases per million per year and LCH is diagnosed most often in
accumulating histiocytes are positive for CD1a or langerin
(CD207) on immunohistochemistry.8 The Birbeck granule is a
Correspondence: Akira Morimoto, MD PhD, Department of Pediatrics, Langerhans cell-specific tennis racquet-shaped organelle that can
Jichi Medical University School of Medicine, 3311-1, Yakushiji,
Shimotsuke, Tochigi 329-0498, Japan. Email: akira@jichi.ac.jp be detected by electron microscopy. In terms of diagnostic value,
Received 19 January 2014; revised 27 March 2014; accepted 1 May the Birbeck granule has been replaced by langerin, which is a
2014. cell-surface receptor that induces the formation of the Birbeck

© 2014 Japan Pediatric Society


452 A Morimoto et al.

granule. Active LCH lesions contain granulomas caused by the Pathogenesis


aggregation of LCH cells as well as a number of various inflam- The putative cell interaction and cytokine /chemokine network
matory cells. In the later stages of LCH, macrophages are more are illustrated in Figure 1.
predominant than LCH cells in the lesions, and xanthomatous
and fibrotic changes are often observed. In cases where the aggre-
gating histiocytes are negative for CD1a staining, juvenile LCH cell characteristics
xanthogranuloma, Erdheim–Chester disease or Rosai–Dorfman Compared to normal Langerhans cells, LCH cells are more
disease may be considered. When the CD1a-positive cells exhibit proliferative and have a lower antigen-presenting capability.
prominent dysmorphic features and mitotic figure, Langerhans This suggests that during their maturation, LCH cells
cell sarcoma should be ruled out.9,10 Characteristics of these types become arrested at an activated state.19 A recent study using
of non-Langerhans cell histiocytosis are summarized in an expression array revealed that LCH cells exhibit a unique
Table 1. transcription profile that separates them not only from
plasmacytoid but also from epidermal Langerhans cells.20,21
Etiology In particular, LCH cells are the only dendritic cells that
It has been debated for many years whether LCH is a neoplasm co-express Notch1 and its Jagged-1 and -2 ligands.21 Interest-
or is reactive in nature. Several lines of evidence suggest that ingly, when hematopoietic progenitor cells are cultured with
LCH is reactive: LCH lesions sometimes regress sponta- granulocyte-macrophage colony-stimulating factor (GM-CSF),
neously,11 some virus DNA sequences have been detected in the presence of fibroblasts expressing Jagged-1 stimulates
peripheral blood and tissues from LCH patients,12 and there is the accumulation of dendritic cell precursors, not mature
no evidence that LCH cells are immortalized. However, the fact dendritic cells, because their transition to terminally differenti-
that LCH cells are monoclonal13,14 and sometimes show chro- ated dendritic cells is blocked.22 Further supporting the possi-
mosomal deletion or gain15 suggests the opposite conclusion. bility that Notch1 may contribute to LCH pathogenesis is
Further supporting this is that an oncogenic BRAF V600E a report of a patient who first developed T-cell acute
mutation was recently discovered in LCH cells;16 this mutation lymphoblastic leukemia and then LCH: the leukemic cells and
is detected in more than half of LCH patients.17 However, the LCH cells of this patient harbored the same activating Notch1
clinical significance of this mutation has not been settled mutation.23
because it occurs equally as frequently in MS and SS LCH, In summary, LCH cells are distinctive immature dendritic
although it was reported recently that BRAF V600E mutation- cells and the JAG-mediated Notch signaling pathway may play
positive patients may have a higher reactivation rate than nega- an important role in maintaining LCH cells in an immature
tive patients.18 state.

Table 1 Characteristics of non-Langerhans cell histiocytoses

Disease Subtype Susceptible age Character of lesions Pathology


Juvenile Cutaneous type (75%) <2 years old Multiple nodular small papules CD1a (–), Langerin (–), S100
xanthogranuloma Soft tissue type (15%) <2 years old Soft tissue mass lesions in (–), Fascin (+), Factor XIIa
subcutaneous fat or muscle (+), CD163 (+)
Systemic type (10%) <6 months old Mass lesions in visceral organ – Touton giant cells (cutaneous
subcutis, central nervous system, type)
bone, liver, spleen, lung and
others
Erdheim–Chester >40 years old Symmetrical sclerotic lesions in the Similar to juvenile
disease long bones. xanthogranuloma
Non-osseous lesions – ureteral
stricture associated with
retroperitoneal fibrosis,
xanthomatous skin lesion, central
diabetes insipidus and pulmonary
fibrosis (50%).
Rosai–Dorfman Children and Painless bilateral cervical CD1a (–), Langerin (–), S100
disease young adults lymphadenopathy (80%) (+), CD163 (+)
Extranodal lesions – osteolytic bone Intracytoplasmic emperipolesis of
lesions, maculopapular skin rash, lymphocytes, neutrophils,
orbital masses and others (50%). plasma cells and red blood
cells to the histiocytes
Langerhans cell Any age Multiple organ involvement – lymph CD1a (+), Langerin (+), S100 (+)
sarcoma nodes, liver, spleen, bone, lung Prominent nuclear pleomorphism
and others and mitoses

© 2014 Japan Pediatric Society


Recent advances in LCH 453

Fig. 1 Putative cell interaction and


cytokine/chemokine network in
Langerhans cell histiocytosis (LCH)
lesions. GM-CSF, granulocyte-macro-
phage colony-stimulating factor; HLA-
DR, human leukocyte antigen-DR;
IL, interleukin; M-CSF, macrophage
colony-stimulating factor; OPG, oste-
oprotegerin; RANKL, receptor activator
for nuclear factor κ B ligand; TNF,
tumor necrosis factor.

Interaction of LCH cells with other cells that infiltrate rather than to their uncontrolled proliferation, and that because
LCH lesions LCH cells bear immature dendritic cell characteristics, they may
LCH lesions not only contain LCH cells, they also bear inflam- promote the expansion of regulatory T cells, which in turn results
matory cells, including T lymphocytes, macrophages, plasma in the failure of the host immune system to eliminate LCH cells.32
cells, eosinophils, osteoclast-like multinucleated giant cells, and In summary, LCH lesions are characterized by a cytokine/
neutrophils. LCH cells also express the immature dendritic cell chemokine storm and interactions between infiltrating cells that
marker CCR6 and produce its ligand (CCL20/MIP-3α) as well play a role in the accumulation of LCH cells and other infiltrating
as CCL5/RANTES (CCR1/3/5 ligand) and CXCL11/I-TAC cells and are responsible for various clinical features of LCH.
(CXCR3 ligand).24 Recently, it was reported that LCH cells from
patients with multifocal lesions express CXCR4 and its ligand Osteoclastogenesis in LCH
(CXCL12/SDF-1).25 These chemokine receptors and their ligand Receptor activator for nuclear factor κ B ligand (RANKL) is
interactions may play a role in the hematogenous migration of expressed by osteoblast/stromal cells and promotes bone resorp-
not only LCH cells but also eosinophils and CD4-positive T cells tion by binding to RANK on osteoclast (OC) precursors, which
into the lesions. These infiltrating cells and LCH cells in turn activates these cells. Osteoprotegerin (OPG), which is also
stimulate each other to produce abundant cytokines, such as secreted by osteoblast/stromal cells, is a decoy receptor of
GM-CSF, interferon (IFN)-γ, tumor necrosis factor (TNF)-α, RANKL. Thus, the balance between RANKL and OPG dictates
interleukin (IL)-1α, IL-2, IL-3, IL-4, IL-5, IL-7, and IL-10.26 OC formation and thereby regulates bone resorption. In LCH,
Moreover, in LCH lesions, LCH cells express CD40 at high OC-like multinucleated giant cells (MGC) are present not only in
levels while T cells express CD154 (the CD40 ligand): interac- the bone but also the skin and lymph node lesions. Moreover,
tions between CD40 and CD154 are essential for the activation of both the LCH cells and T cells in these lesions produce the
both the antigen-presenting cells and the T cells.27 We also found OC-inducing cytokines of RANKL and macrophage colony-
that the serum levels of lesional inflammatory cytokines/ stimulating factor (M-CSF).33 Enzymes derived from OC-like
chemokines and dendritic cell/macrophage-activating factors, MGC may play a major role in the chronic tissue destruction that
such as IL-2R, IL-12p40, IL-18 and CXCL9, are markers of is seen in LCH lesions.33 We found that patients with SS LCH and
disease dissemination and severity.28 Recently, it was reported multiple bone lesions had significantly higher soluble RANKL/
that LCH cells also abundantly express the pleiotropic cytokine OPG ratios than patients with MS LCH; moreover, the soluble
osteopontin (OPN).20 Moreover, bronchoalveolar lavage cells in RANKL/OPG ratios correlated positively with the number of
patients with pulmonary LCH have upregulated OPN expression bone lesions.34
and overexpression of OPN in rat lungs induces lesions similar to Coury et al. showed that patients with active LCH have high
pulmonary LCH.29 OPN has a variety of functions that include serum levels of IL-17A, which is mainly produced by LCH
promoting the generation of T helper (Th)1 and Th17 cells, cells.35 In vitro, the serum IL-17A leads to a fusion between
recruiting histiocytes/monocytes, and activating osteoclasts.30 We immature dendritic cells that results in the formation of OC-like
found that patients with MS LCH have much higher serum OPN MGC. This IL-17A-dependent fusion activity of the serum cor-
levels than patients with SS LCH.31 Finally, a recent study sug- relates with LCH activity. An IL-17A autocrine model for LCH
gests that the accumulation of LCH cells is due to their survival was suggested. We also found that patients with LCH have higher

© 2014 Japan Pediatric Society


454 A Morimoto et al.

serum levels of IL-17A than controls, but that the serum IL-17A Bone lesions
levels of patients with MS LCH and SS LCH did not differ The bone is involved in about 80% of patients with LCH.40 In
significantly. However, we also observed that LCH cells in MS pediatric patients, the skull (Fig. 3a) is the most often affected
disease had higher levels of IL-17A receptor (IL-17RA) protein location followed by the spine (Fig. 3b), extremities, pelvic
expression than those in SS disease and that IL-17RA expression bone, and ribs.41 In adult patients, the most affected bone is the
levels helped to distinguish between LCH subclasses.36 In addi- jaw.5 X-rays typically demonstrate osteolytic “punched out”
tion, we found that the cleaved form of OPN plays a critical role lesions with sharp margins. Fluorodeoxyglucose-positron emis-
in driving immature dendritic cell differentiation into OC-like sion tomography42 and whole-body magnetic resonance
MGC in an autocrine and/or paracrine fashion.37 imaging (MRI)43 are also useful for detecting bone lesions and
These data suggest that the T cells and LCH cells in LCH evaluating their response to treatment. Bone lesions may be
lesions interact and produce abundant amounts of cytokines that asymptomatic or accompanied with pain and soft tissue
induces the differentiation and activation of OC, which in turn swelling. When LCH is localized solely to the bone, the clinical
produces large amounts of tissue-destructive enzymes. course is generally benign and it sometimes resolves sponta-
Clinical manifestations neously over a period of months to years. However, it can
also result in critical or irreversible symptoms, such as visual
As LCH affects a number of different organs, its clinical signs loss or exophthalmos (due to orbital involvement), conductive
and symptoms can be extremely variable. In most pediatric hearing loss (due to mastoid antrum lesions), loss of teeth
patients with SS LCH, lesions occur in the bone with single-site (due to jaw diseases), and spinal paralysis (due to vertebral
or multifocal involvement, although the lesions can also occur in lesions).
the skin.38 By contrast, in adults with SS LCH, the lung is the
most common site of lesions.5 In patients with MS LCH,
the organs involved are mainly the bone and skin, followed by the Skin lesions
hematopoietic system, lymph node, liver, spleen, soft tissue, Skin involvement is seen in approximately half of all patients and
lung, thymus, pituitary gland, and other organs.39 The most particularly frequent in infants. These patients present with
common initial manifestations are the development of a soft various lesions, including erythema, papules, nodules, petechiae,
tissue mass, bone pain, skin rash, fever, otorrhea, and lymphad- vesicles, crusted plaques and seborrhea-like eruptions. Ulcerative
enopathy. There are no specific laboratory markers for LCH. In lesions in the genital or inguinal region may also be present.44 It
many cases, LCH presents with nonspecific inflammatory signs is essential that patients with skin lesions undergo evaluations of
that arise from chronic inflammation. A diagnostic approach is other viscera because most of these patients have MS disease. In
summarized in Figure 2. neonatal infants, isolated skin lesions can regress spontaneously

Fig. 2 Diagnosis approach of


Langerhans cell histiocytosis (LCH).
CDI, central diabetes insipidus; CNS,
central nervous system; CT, computed
tomography; MRI, magnetic resonance
imaging; MS, multisystem.

© 2014 Japan Pediatric Society


Recent advances in LCH 455

Fig. 3 Radiological findings of


Langerhans cell histiocytosis patients.
(a) Computed tomography (CT) of a
skull lesion. Both inner and outer plates
are affected. (b) Magnetic resonance
imaging (MRI) (T1-weighted image) of
a vertebral lesion (kindly provided by
Dr A. Mababe, St Luke’s International
Hospital). Vertebra body of Th9 is
collapsed and extensive paravertebral
soft tissue is present. (c) CT of
pulmonary lesions (kindly provided by
Dr M. Hiwatari, University of Tokyo).
Multiple cysts show honeycomb appear-
ance. (d) MRI (T1-weighted image) of a
pituitary lesion. White arrow indicates
thickened pituitary stalk and black
arrow indicates loss of bright spot in
posterior pituitary.

over weeks to months.45 However, some infants with isolated skin with thrombocytopenia, with or without leukopenia and
lesions at diagnosis can develop MS disease followed by a fatal hypoalbuminemia, are associated with a worse prognosis in
clinical course.46 children with MS LCH.50 It is considered to constitute risk-
organ involvement51 (Table 2).
Lung lesions
In children, pulmonary involvement is usually part of MS Liver and spleen lesions
disease. It has been shown that pulmonary LCH in adults gener- Liver and/or spleen involvement occurs in 20% of patients39 and
ally occurs in cigarette smokers and frequently regresses after the is characterized by organomegaly and/or liver dysfunction
cessation of smoking.47 The lung pathology is associated with (Table 2).51 Histological examination of the liver indicates portal
cough, dyspnea, pleural effusion, and recurring pneumothorax. infiltrates that can induce bile duct destruction and periportal
High resolution-computed tomography may reveal reticular or fibrosis (sclerosing cholangitis), leading to biliary cirrhosis with
micronodular opacities as well as large nodules and honeycomb- portal hypertension and ultimately secondary hypersplenism.
ing (Fig. 3c). KL-6 may be a useful marker for pulmonary
Other lesions
involvement.48 In children with MS LCH, pulmonary involve-
ment is not an independent prognostic variable.49 Oral mucosa infiltration may appear as ulcerations or swelling of
gingiva.52 Infiltration of the gastrointestinal tracts sometimes
Hematopoietic involvement occurs and can cause vomiting, abdominal pain, constipation,
Hematopoietic involvement is seen in disseminated LCH. In intractable diarrhea with blood and/or mucus53 and malabsorption
severe cases, serious anemia and thrombocytopenia may of nutrients.54 The lymph nodes that are most commonly affected
develop and are often associated with a secondary are those in the cervical, axillary and inguinal areas.55 Rarely, the
hemophagocytic syndrome followed by a fatal course. Anemia nodes can become massive and cause upper airway obstruction.

© 2014 Japan Pediatric Society


456 A Morimoto et al.

Table 2 Definition of risk-organ involvement† temic chemotherapy effectively reduces its development.60 About
half of the patients with CDI develop anterior pituitary hormone
Hematopoietic involvement (with or without bone-marrow
involvement)
deficiencies and/or ND-CNS disease during follow up, usually
At least two of the following: after a disease course of several years.62 LCH involvement of the
• Anemia: hemoglobin <10 g/dL, infants <9 g/dL (not due to other pituitary gland can be shown by MRI findings, namely, pituitary
causes, e.g. iron deficiency) stalk thickening and loss of the physiological high-intensity
• Leukocytopenia: leukocytes <4 000 /mm3 signal of the posterior pituitary lobe on T1-weighted images
• Thrombocytopenia: platelets <100 000 /mm3
Liver involvement
(Fig. 3d). Patients with hypothalamic-pituitary lesions may
• Enlargement >3 cm below the costal margin in the midclavicular present with non-endocrine hypothalamic dysfunction, such as
line eating disorder, thirst, fatigue, autonomic disturbance, tempera-
and/or ture instability, memory deficit, and disturbed consciousness.63
• Liver dysfunction (i.e. hypoproteinemia <5.5 g/dL, The hypothalamic lesions can be indicated by MRI as
hypoalbuminemia <2.5 g/dL, not due to other causes) gadolinium-enhanced masses.64
and/or
• Histopathological diagnosis
Spleen involvement Neurodegenerative central nervous system disease
• Enlargement >2 cm below the costal margin at the midclavicular ND-CNS disease may develop over the years after the onset of
line
Lung involvement
the disease, often when the disease is considered quiescent.65,66
• Typical changes on HR-CT (low dose multidetector CT if Three years after the diagnosis of LCH, more than half of patients
available) with CNS-risk lesions and/or CDI demonstrate bilateral symmet-
and/or rical lesions in the cerebellar white matter and basal ganglia
• Histopathological / cytological diagnosis (radiological ND-CNS disease) in MRI studies using

Definition by the Histiocyte Society in 2009.51 T2-weighted or fluid-attenuated inversion recovery images.67
These brain MRI abnormalities gradually deteriorate and never
reverse. One quarter of patients with the brain MRI abnormalities
Thymic involvement mainly occurs in young children and is will present with neurological symptoms (neurological ND-CNS
diagnosed when CT indicates thymus enlargement and calcifica- disease), including ataxia, tremor, dysarthria, dysphagia and
tions.56 The thyroid gland,57 pancreas, and kidney58 are also occa- hyperreflexia, within several years, followed by further deterio-
sionally involved. ration. Patients with neurological ND-CNS disease eventually
become bedridden. Histologically, ND-CNS disease is character-
Hypothalamic-pituitary disease
ized by the presence of CD8-positive T-lymphocyte infiltration,
In 25% of all patients with LCH and 50% of patients with MS microglia activation, gliosis, and neuronal and axonal destruction
disease, infiltration and dysfunction of the pituitary gland and/or with secondary demyelination, as is observed in autoimmune
adjacent hypothalamus is observed.59 The most frequent manifes- encephalitis. CD1a-positive LCH cells may be absent.68 First-line
tation is CDI, which may precede, co-occur, or follow other therapies for LCH that include cytarabine (Ara-C) may reduce
symptoms and signs of the disease. The cumulative incidence of the incidence of ND-LCH.69,70 Currently, there is no established
CDI continues to increase, even 10 years after the diagnosis of therapy for ND-CNS disease, although there is some promising
LCH.60 CDI occurs more often in patients with craniofacial bone evidence that intravenous immunoglobulin71,72 or Ara-C73 treat-
lesions (with the exception of vault), ear, eye and/or oral lesions61 ment may prevent the progression of ND-CNS disease.
and thus these lesions are known as CNS-risk lesions (Table 3).
Systemic chemotherapy rarely cures CDI but appropriate sys- Treatment and outcome
The clinical course of LCH varies quite widely depending on the
Table 3 Definition of CNS-risk lesions† extent of organ involvement. The mortality rate of patients with
SS disease or without risk-organ involvement is less than 5%.
Craniofacial bone involvement However, children with MS disease and risk-organ involvement
Lesions in the orbital, temporal, mastoid, sphenoidal, zygomatic, or
ethmoidal bones; the maxilla or paranasal sinuses; or cranial (Table 2) have a worse prognosis: these children often have fatal
fossa; with intracranial soft tissue extension outcomes despite intensive treatment and their mortality is
Ear involvement reported to be 10–50%.39 Treatment of LCH should be planned
External otitis, otitis media, otorrhea; or lesions in the temporal according to the clinical presentation and the extent of organ
bone, mastoid, or petrous bone involvement. In 2010, the Infant LCH study group of Research on
Eye involvement
Proptosis, exophthalmos, or lesions in the orbits; zygomatic or Measures for Intractable Diseases by the Ministry of Health
sphenoidal bone Labour and Welfare in Japan presented the treatment guidance
Oral involvement for childhood LCH (Fig. 4). Etoposide (VP-16) is no longer
Lesions in the oral mucosa, gums, palatal bone, maxilla, and considered to be a reasonable therapeutic agent because it is not
mandible more efficacious than vinblastine74 and has been shown to cause

Definition by the Histiocyte Society in 2009.51 therapy-related acute myeloid leukemia (t-AML).75 Radiation is

© 2014 Japan Pediatric Society


Recent advances in LCH 457

Fig. 4 Treatment guideline for newly


diagnosed childhood Langerhans cell
histiocytosis (LCH). Modification of the
guidelines produced by the Infant LCH
study group of Research on Measures
for Intractable Diseases by the Ministry
of Health Labour and Welfare in Japan
in 2010. 2-CdA, cladribine; Ara-C,
cytarabine; CNS-risk lesion, central
nervous system risk lesion (see Table 3);
MS, multisystem; PSL, prednisolone;
RIC-SCH, reduced-intensity condition-
ing stem cell transplantation; RO, risk
organ involvement (see Table 2);
SS, single-system; VCR, vincristine.

rarely used in children because of the reported increased risk of yields more favorable outcomes for risk organ-positive patients:
secondary malignancies, particularly brain tumors, in irradiated their mortality rate is about 10%.80,81 A major prognostic factor is
areas.76 a response to the first 6 weeks of systemic multi-agent chemo-
therapy: risk organ-positive patients who exhibit progressive
Single-system LCH disease during combination chemotherapy have an extremely
In SS LCH, the main aims of treatment are to lessen symptoms high mortality rate of between 40% and 80%.78–83 Moreover,
and reduce the chance of permanent sequelae. In patients with despite being treated with the regimens described above, the
localized, unifocal bone disease, LCH may resolve sponta- reactivation rate of patients with MS disease remains high (up to
neously. In the case of a single bone lesion without symptoms, a 30%).79,81 To reduce the reactivation rate, the JLSG-02 regimen
wait-and-see approach or diagnostic curettage is the standard plus intensification via dose-up of VCR in the early maintenance
method of care. Local injection of corticosteroid may be used for phase was developed. The ongoing LCH-12 trial, which was
these bone lesions.77 Patients with CNS-risk lesions or multifocal initiated in Japan recently, is testing the efficacy of this regimen.
bone disease should receive systemic chemotherapy with vinca
alkaloids and corticosteroids for 6 or 12 months to prevent the Adult LCH
development of late sequelae, such as CDI. Radical operation of In adults, lung disease can be a life-threatening complication: one
large bone lesions is discouraged as this often results in disfig- study showed that 5 years after the diagnosis, patients with lung
urement and loss of bone. When there is skin involvement only, LCH lesions have a mortality rate of approximately 25%.84 In
the best option is a wait-and-see approach. Alternatively, patients patients with isolated pulmonary LCH with functional impair-
can be provided therapies such as topical corticosteroids. A ment, systemic chemotherapy may be indicated to reduce further
nationwide survey in Japan suggests that appropriate chemo- parenchymal destruction, although it remains unclear whether
therapy for patients with a single bone lesion results in an excel- chemotherapy reduces the mortality rate.
lent prognosis.38 Systemic chemotherapy is required for multifocal SS or MS
disease also in adults.85 It should be noted that although most
Multi-system LCH generally have indolent and chronic clinical courses, some adult
In MS LCH, the main aims of treatment are to increase survival LCH patients exhibit rapid progression and fatal courses.86
and to reduce the incidence of late sequelae. The most commonly However, the optimal systemic therapy for adult LCH has not yet
used regimen in other countries is systemic chemotherapy with been developed.87–90 Multifocal adult LCH is an orphan disease.
vinblastine (VBL) and corticosteroid for 6–12 months. Three This has hampered the development of an effective systemic
clinical trials designated as LCH-I, -II, and -III have tested this therapy for several reasons, as follows. First, it often takes time to
regimen74,78,79 and showed that it yields excellent survival rates in correctly diagnose LCH in adults. Second, it is very common in
risk organ-negative patients, even in infants.78,79 However, despite adult LCH to adopt a “wait and see” strategy after diagnosis, even
this treatment, the mortality rates of risk organ-positive patients when it is multifocal LCH. Third, the wide range of symptoms of
remain high at 16–38%.74,78,79 The combination chemotherapy this rare disease causes patients to visit a variety of clinics that
consisting of Ara-C, vincristine (VCR), and prednisolone that apply various therapeutic regimens. Moreover, adult patients are
was introduced by the Japan LCH Study Group (JLSG-96/02) often reluctant to take a leave of absence from their jobs for

© 2014 Japan Pediatric Society


458 A Morimoto et al.

hospitalization. For this reason, the JLSG has formulated an hearing loss, neurological problems, growth-hormone deficiency,
ambulatory treatment regimen for adult patients with mul- loss of teeth, pulmonary fibrosis, and biliary cirrhosis with portal
tifocal SS or MS disease that consists of combinations of hypertension. The most serious of these sequelae are those in the
VBL/prednisolone (PSL) and methotrexate with daily CNS, such as CDI and ND-CNS disease; these sequelae occur
6-mercaptopurine (denoted the Special C regimen). This regimen particularly frequently in MS disease.59,70 The incidence of these
has shown signs of efficacy, particularly in patients with sequelae increases over follow-up. Further novel therapeutic
multifocal SS disease.90 Adult LCH may also respond to Ara-C, measures are required to reduce these permanent sequelae.
which is an effective agent for pediatric LCH.89 JLSG is now
Conclusions
planning a clinical trial for adult multifocal LCH using the
Special C regimen as the first-line treatment and Ara-C/PSL as The pathogenesis of LCH remains obscure and the optimal treat-
the salvage therapy for poor responders. ment for LCH has not been established, although incremental
progress has been made. Whether LCH is a reactive or neoplas-
Salvage therapy and new treatment tic disorder remains unclear. Proinflammatory cytokines/
In patients who have risk-organ involvement and do not respond chemokines are known to participate in LCH, which suggests that
to the first-line therapy, more aggressive salvage therapy should LCH is an immune disorder. However, more than half of LCH
be considered. Myeloablative therapy using a combination of patients have the oncogenic BRAF mutation, which suggests that
high-dose Ara-C and cladribine (2-CdA)91,92 and the single agent LCH is a neoplastic disorder. There are several major issues in
clofarabine93 have been reported to be effective for these patients. the treatment of LCH that remain to be resolved. First, it remains
However, profound and prolonged myelosuppression should be unclear how to rescue patients with risk-organ involvement who
anticipated in these therapies. Recently, it was reported that when do not respond to first-line therapy. Moreover, the optimal treat-
patients with severe and refractory BRAF V600E-positive LCH ment for multifocal adult LCH is not known. Third, protocols that
were treated with vemurafenib, a BRAF inhibitor that was newly effectively reduce and treat CNS-related consequences are
approved for melanoma by the US Food and Drug Administra- needed. To elucidate the pathogenesis of LCH and develop the
tion, they showed substantial and rapid clinical and biological new treatment, genome-wide association studies107 and genera-
improvements without severe toxicities or adverse events.94 tion of NOG mice model108 should be performed in the future.
Allogeneic hematopoietic stem cell transplantation is another
promising approach as a salvage therapy for children with refrac- Acknowledgment
tory LCH because of its immunomodulatory effect.95 A reduced- We thank Dr Shinsaku Imashuku for laying the foundation of the
intensity conditioning regimen may be sufficient to induce a cure, JLSG studies, the members of JLSG for providing patient infor-
which would decrease the treatment-related mortality.96 mation, and Ms Yasuko Hashimoto for her excellent secretarial
In patients who show reactivation in non-risk organs while assistance. This work was supported by a Grant for Research on
they are off therapy, treatment with pamidronate combined with Measures for Intractable Diseases from the Ministry of Health,
Cox2 inhibitor97 or 2-CdA monotherapy98,99 may be effective. Labor and Welfare, Japan.
Careful attention is required to prevent known adverse effects of
therapy with 2-CdA, namely, the development of severe persis- References
tent bone-marrow failure and myelodysplastic syndrome.100,101 1 Salotti JA, Nanduri V, Pearce MS, Parker L, Lynn R, Windebank
In addition, liver or lung transplants have been successfully KP. Incidence and clinical features of Langerhans cell
performed in patients with end-stage organ involvement of histiocytosis in the UK and Ireland. Arch. Dis. Child. 2009; 94:
376–80.
LCH.102,103
2 Guyot-Goubin A, Donadieu J, Barkaoui M, Bellec S, Thomas C,
Clavel J. Descriptive epidemiology of childhood Langerhans cell
Reactivation and permanent sequelae histiocytosis in France, 2000–2004. Pediatr. Blood Cancer 2008;
Nearly half of patients with MS disease exhibit reactivation, 51: 71–5.
which occurs in an unpredictable manner.104,105 Some patients 3 Horibe K, Saito AM, Takimoto T et al. Incidence and survival
rates of hematological malignancies in Japanese children and
experience reactivation several times. Most of the first reactiva- adolescents (2006–2010): based on registry data from the Japa-
tions occur within 2 years after diagnosis. Reactivations occur nese Society of Pediatric Hematology. Int. J. Hematol. 2013; 98:
most frequently in bone and are rare in risk organs. Reactivated 74–88.
lesions may sometimes resolve spontaneously. Most of the 4 Stockschlaeder M, Sucker C. Adult Langerhans cell histiocytosis.
patients with reactivation undergo second disease resolution and Eur. J. Haematol. 2006; 76: 363–8.
5 Aricò M, Girschikofsky M, Généreau T et al. Langerhans cell
survive. However, they have an increased risk of permanent histiocytosis in adults. Report from the International Registry of
sequelae. the Histiocyte Society. Eur. J. Cancer 2003; 39: 2341–8.
In many LCH patients, permanent sequelae are common.106 6 Aricò M, Danesino C. Langerhans’ cell histiocytosis: is there a
They are most often the result of the infiltrative nature of the role for genetics? Haematologica 2001; 86: 1009–14.
disease itself, which causes tissue destruction and granulomatous 7 Egeler RM, Neglia JP, Aricò M et al. The relation of Langerhans
cell histiocytosis to acute leukemia, lymphomas, and other solid
fibrosis or gliosis of various tissues. Seventy percent of patients tumors. The LCH-Malignancy Study Group of the Histiocyte
with MS disease and 25% of SS disease patients develop one or Society. Hematol. Oncol. Clin. North Am. 1998; 12: 369–
more life-long sequelae, including CDI, orthopedic problems, 78.

© 2014 Japan Pediatric Society


Recent advances in LCH 459

8 Jaffe R, Weiss LM, Facchetti F. Tumours derived from 27 Egeler RM, Favara BE, Laman JD, Claassen E. Abundant expres-
Langerhans cells. In: Swerdlow SH, Campo E, Harris NL et al. sion of CD40 and CD40-ligand (CD154) in paediatric
(eds). WHO Classification of Tumours, Tumours of Langerhans cell histiocytosis lesions. Eur. J. Cancer 2000; 36:
Haematopoietic and Lymphoid Tissues, 4th edn. IARC, Lyon, 2105–10.
2008; 358–60. 28 Morimoto A, Nakamura S, Shioda Y et al. Comprehensive analy-
9 Lau SK, Chu PG, Weiss LM. Immunohistochemical expression of sis of serum cytokine/ chemokine and growth factors in pediatric
Langerin in Langerhans cell histiocytosis and non-Langerhans patients with Langerhans cell histiocytosis. Pediatr. Blood
cell histiocytic disorders. Am. J. Surg. Pathol. 2008; 32: 615–9. Cancer 2011; 56: 696. [abstract].
10 Weitzman S, Whitlock J. Uncommon histiocytic disorder: the 29 Prasse A, Stahl M, Schulz G et al. Essential role of osteopontin in
non-Langerhans cell histiocytoses. In: Weitzman S, Egeler RM smoking-related interstitial lung diseases. Am. J. Pathol. 2009;
(eds). Histiocytic Disorders of Children and Adults. Cambridge 174: 1683–91.
University press, Cambridge, 2005; 293–320. 30 Uede T. Osteopontin, intrinsic tissue regulator of intractable
11 Minkov M, Prosch H, Steiner M et al. Langerhans cell inflammatory diseases. Pathol. Int. 2011; 61: 265–80.
histiocytosis in neonates. Pediatr. Blood Cancer 2005; 45: 802–7. 31 Oh Y, Morimoto A, Shioda Y, Imamura T, Kudo K, Imashuku S.
12 Murakami I, Matsushita M, Iwasaki T et al. Merkel cell High serum osteopontin levels in pediatric patients with high risk
polyomavirus DNA sequences in peripheral blood and tissues Langerhans cell histiocytosis. Cytokine. 2014 Jul 30. doi:
from patients with Langerhans cell histiocytosis. Hum. Pathol. 10.1016/j.cyto.2014.07.002. [Epub ahead of print].
2014; 45: 119–26. 32 Senechal B, Elain G, Jeziorski E et al. Expansion of regulatory T
13 Willman CL, Busque L, Griffith BB et al. Langerhans’-cell cells in patients with Langerhans cell histiocytosis. PLoS Med.
histiocytosis (histiocytosis X) – a clonal proliferative disease. 2007; 4: e253.
N. Engl. J. Med. 1994; 331: 154–60. 33 da Costa CE, Annels NE, Faaij CM, Forsyth RG, Hogendoorn
14 Yu RC, Chu C, Buluwela L, Chu AC. Clonal proliferation of PC, Egeler RM. Presence of osteoclast-like multinucleated giant
Langerhans cells in Langerhans cell histiocytosis. Lancet 1994; cells in the bone and nonostotic lesions of Langerhans cell
343: 767–8. histiocytosis. J. Exp. Med. 2005; 201: 687–93.
15 Murakami I, Gogusev J, Fournet JC, Glorion C, Jaubert F. Detec- 34 Ishii R, Morimoto A, Ikushima S et al. High serum values of
tion of molecular cytogenetic aberrations in Langerhans cell soluble CD154, IL-2 receptor, RANKL and osteoprotegerin in
histiocytosis of bone. Hum. Pathol. 2002; 33: 555–60. Langerhans cell histiocytosis. Pediatr. Blood Cancer 2006; 47:
16 Badalian-Very G, Vergilio JA, Degar BA et al. Recurrent BRAF 194–9.
mutations in Langerhans cell histiocytosis. Blood 2010; 116: 35 Coury F, Annels N, Rivollier A et al. Langerhans cell
1919–23. histiocytosis reveals a new IL-17A-dependent pathway of
17 Satoh T, Smith A, Sarde A et al. B-RAF mutant alleles associated dendritic cell fusion. Nat. Med. 2008; 14: 81–7.
with Langerhans cell histiocytosis, a granulomatous pediatric 36 Murakami I, Morimoto A, Oka T et al. IL-17A receptor expres-
disease. PLoS ONE 2012; 7: e33891. sion differs between subclasses of Langerhans cell histiocytosis,
18 Berres ML, Lim KP, Peters T et al. BRAF-V600E expression in which might settle the IL-17A controversy. Virchows Arch. 2013;
precursor versus differentiated dendritic cells defines clinically 462: 219–28.
distinct LCH risk groups. J. Exp. Med. 2014; 211: 669–83. 37 Oh Y, Oh I, Morimoto J et al. Osteopontin has a crucial role in
19 Geissmann F, Geissmann F, Lepelletier Y, Fraitag S et al. Dif- osteoclast-like multinucleated giant cell formation. J. Cell.
ferentiation of Langerhans cells in Langerhans cell histiocytosis. Biochem. 2014; 115: 85–95.
Blood 2001; 97: 1241–8. 38 Morimoto A, Ishida Y, Suzuki N et al. Nationwide survey of
20 Allen CE, Li L, Peters TL et al. Cell-specific gene expression in single-system single site LCH in Japan. Pediatr. Blood Cancer
Langerhans cell histiocytosis lesions reveals a distinct profile 2010; 54: 98–102.
compared with epidermal Langerhans cells. J. Immunol. 2010; 39 Donadieu J, Egeler RM, Pritchard J. Langerhans cell
184: 4557–67. histiocytosis: a clinical update. In: Weitzman S, Egeler RM (eds).
21 Hutter C, Kauer M, Simonitsch-Klupp I et al. Notch is active in Histiocytic Disorders of Children and Adults. Cambridge Univer-
Langerhans cell histiocytosis and confers pathognomonic features sity Press, Cambridge, 2005; 95–129.
on dendritic cells. Blood 2012; 120: 5199–208. 40 Weitzman S, Egeler RM. Langerhans cell histiocytosis of bone.
22 Cheng P, Nefedova Y, Corzo CA, Gabrilovich DI. Regulation of In: Weitzman S, Egeler RM (eds). Histiocytic Disorders of Chil-
dendritic-cell differentiation by bone marrow stroma via different dren and Adults. Cambridge University Press, Cambridge, 2005;
Notch ligands. Blood 2007; 109: 507–15. 154–73.
23 Rodig SJ, Payne EG, Degar BA et al. Aggressive Langerhans cell 41 Imashuku S, Kinugawa N, Matsuzaki A et al. Langerhans cell
histiocytosis following T-ALL: clonally related neoplasms with histiocytosis with multifocal bone lesions: comparative clinical
persistent expression of constitutively active NOTCH1. Am. features between single and multi-systems. Int. J. Hematol. 2009;
J. Hematol. 2008; 83: 116–21. 90: 506–12.
24 Annels NE, Da Costa CE, Prins FA, Willemze A, Hogendoorn 42 Phillips M, Allen C, Gerson P, McClain K. Comparison of FDG-
PC, Egeler RM. Aberrant chemokine receptor expression and PET scans to conventional radiography and bone scans in man-
chemokine production by Langerhans cells underlies the patho- agement of Langerhans cell histiocytosis. Pediatr. Blood Cancer
genesis of Langerhans cell histiocytosis. J. Exp. Med. 2003; 197: 2009; 52: 97–101.
1385–90. 43 Goo HW, Yang DH, Ra YS et al. Whole-body MRI of
25 Quispel W, Stegehuis-Kamp J, Santos-Boeij S, van den Bos C, Langerhans cell histiocytosis: comparison with radio-
van Halteren A, Egeler RM. CXCR4 expression by Langerhans graphy and bone scintigraphy. Pediatr. Radiol. 2006; 36: 1019–
cell histiocytosis cells is associated with disease outcome. 29th 31.
Annual Meeting of The Histiocyte Society. 2013, [abstract]. 44 Krafchik B, Pope E, Walsh S. Histiocytosis of the skin in children
26 Egeler RM, Favara BE, van Meurs M, Laman JD, Claassen E. and adults. In: Weitzman S, Egeler RM (eds). Histiocytic Disor-
Differential in situ cytokine profiles of Langerhans-like cells and ders of Children and Adults. Cambridge University Press, Cam-
T cells in Langerhans cell histiocytosis: abundant expression of bridge, 2005; 130–53.
cytokines relevant to disease and treatment. Blood 1999; 94: 45 Battistella M, Fraitag S, Teillac DH, Brousse N, de Prost Y,
4195–201. Bodemer C. Neonatal and early infantile cutaneous Langerhans

© 2014 Japan Pediatric Society


460 A Morimoto et al.

cell histiocytosis: comparison of self-regressive and non-self- 65 Imashuku S, Ishida S, Koike K et al. Cerebellar ataxia in pediatric
regressive forms. Arch. Dermatol. 2010; 146: 149–56. patients with Langerhans cell histiocytosis. J. Pediatr. Hematol.
46 Lau L, Krafchik B, Trebo MM, Weitzman S. Cutaneous Oncol 2004; 26: 735–9.
Langerhans cell histiocytosis in children under one year. Pediatr. 66 Wnorowski M, Prosch H, Prayer D, Janssen G, Gadner H, Grois
Blood Cancer 2006; 46: 66–71. N. Pattern and course of neurodegeneration in Langerhans cell
47 Suri HS, Yi ES, Nowakowski GS, Vassallo R. Pulmonary histiocytosis. J. Pediatr. 2008; 153: 127–32.
Langerhans cell histiocytosis. Orphanet J. Rare Dis. 2012; 7: 16. 67 Prosch H, Grois N, Wnorowski M, Steiner M, Prayer D. Long-
48 Matsubayashi T, Miwa Y, Saito I, Matsubayashi R. KL-6: marker term MR imaging course of neurodegenerative Langerhans cell
for pulmonary involvement in Langerhans cell histiocytosis in histiocytosis. AJNR Am. J. Neuroradiol. 2007; 28: 1022–8.
infants. J. Pediatr. Hematol. Oncol 2004; 26: 584–6. 68 Grois N, Prayer D, Prosch H, Lassmann H. Neuropathology of
49 Ronceray L, Pötschger U, Janka G, Gadner H, Minkov M. Pul- CNS disease in Langerhans cell histiocytosis. Brain 2005; 128 (Pt
monary involvement in pediatric-onset multisystem Langerhans 4): 829–38.
cell histiocytosis: effect on course and outcome. J. Pediatr. 2012; 69 Imashuku S, Shioda Y, Tsunematsu Y, Imamura T, Morimoto A.
161: 129–33.e1-3. VCR/AraC chemotherapy and ND-CNS-LCH. Pediatr. Blood
50 Braier JL, Rosso D, Latella A et al. Importance of multi-lineage Cancer 2010; 55: 215–6.
hematologic involvement and hypoalbuminemia at diagnosis in 70 Shioda Y, Morimoto A, Imamura T, Kudo K, Imashuku S. Analy-
patients with “risk-organ” multi-system Langerhans cell sis of permanent consequences in pediatric patients with
histiocytosis. J. Pediatr. Hematol. Oncol 2010; 32: e122–5. Langerhans cell histiocytosis: data of the JLSG-96 and JLSG-02
51 Histiocyte Society. Langerhans cell histiocytosis: evaluation and studies in Japan. 29th Annual Meeting of The Histiocyte Society.
treatment guidelines, 2009. [Cited 15 April 2009.] Available from 2013, [abstract].
URL: http://www.histiocytesociety.org/document.doc?id=290. 71 Imashuku S, Shioda Y, Kobayashi R et al. Neurodegenerative
52 Madrigal-Martínez-Pereda C, Guerrero-Rodríguez V, central nervous system disease as late sequelae of Langerhans cell
Guisado-Moya B et al. Langerhans cell histiocytosis: literature histiocytosis. Report from the Japan LCH Study Group.
review and descriptive analysis of oral manifestations. Med. Oral Haematologica 2008; 93: 615–8.
Patol. Oral Cir. Bucal 2009; 14: E222–8. 72 Imashuku S, Okazaki N, Nakayama M et al. Treatment of
53 Yadav SP, Kharya G, Mohan N et al. Langerhans cell neurodegenerative CNS disease in Langerhans cell histiocytosis
histiocytosis with digestive tract involvement. Pediatr. Blood with a combination of intravenous immunoglobulin and chemo-
Cancer 2010; 55: 748–53. therapy. Pediatr. Blood Cancer 2008; 50: 308–11.
54 Shima H, Takahashi T, Shimada H. Protein-losing enteropathy 73 Allen CE, Flores R, Rauch R et al. Neurodegenerative central
caused by gastrointestinal tract-involved Langerhans cell nervous system Langerhans cell histiocytosis and coincident
histiocytosis. Pediatrics 2010; 125: e426–32. hydrocephalus treated with vincristine/cytosine arabinoside.
55 Edelweiss M, Medeiros LJ, Suster S, Moran CA. Lymph node Pediatr. Blood Cancer 2010; 54: 416–23.
involvement by Langerhans cell histiocytosis: a clinicopathologic 74 Gadner H, Grois N, Arico M et al. A randomized trial of treatment
and immunohistochemical study of 20 cases. Hum. Pathol. 2007; for multisystem Langerhans’ cell histiocytosis. J. Pediatr. 2001;
38: 1463–9. 138: 728–34.
56 Ducassou S, Seyrig F, Thomas C et al. Thymus and mediastinal 75 Kudo K, Yoshida H, Kiyoi H, Numata S, Horibe K, Naoe T.
node involvement in childhood Langerhans cell histiocytosis: Etoposide-related acute promyelocytic leukemia. Leukemia 1998;
long-term follow-up from the French national cohort. Pediatr. 12: 1171–5.
Blood Cancer 2013; 60: 1759–65. 76 Haupt R, Nanduri V, Egeler RM. Late effects of Langerhans cell
57 Burnett A, Carney D, Mukhopadhyay S, Scalzetti EM, Leino D, histiocytosis and its association with malignancy. In: Weitzman S,
Souid AK. Thyroid involvement with Langerhans cell Egeler RM (eds). Histiocytic Disorders of Children and Adults.
histiocytosis in a 3-year-old male. Pediatr. Blood Cancer 2008; Cambridge University Press, Cambridge, 2005; 272–92.
50: 726–7. 77 Egeler RM, Thompson RC Jr, Voûte PA, Nesbit ME Jr.
58 Goyal R, Das A, Nijhawan R, Bansal D, Marwaha RK. Intralesional infiltration of corticosteroids in localized
Langerhans cell histiocytosis infiltration into pancreas and Langerhans’ cell histiocytosis. J. Pediatr. Orthop. 1992; 12:
kidney. Pediatr. Blood Cancer 2007; 49: 748–50. 811–4.
59 Grois N, Fahrner B, Arceci RJ et al. Central nervous system 78 Gadner H, Grois N, Pötschger U et al. Improved outcome in
disease in Langerhans cell histiocytosis. J. Pediatr. 2010; 156: multisystem Langerhans cell histiocytosis is associated with
873–81. therapy intensification. Blood 2008; 111: 2556–62.
60 Shioda Y, Adachi S, Imashuku S, Kudo K, Imamura T, Morimoto 79 Gadner H, Minkov M, Grois N et al. Therapy prolongation
A. Analysis of 43 cases of Langerhans cell histiocytosis (LCH)- improves outcome in multisystem Langerhans cell histiocytosis.
induced central diabetes insipidus registered in the JLSG-96 and Blood 2013; 121: 5006–14.
JLSG-02 studies in Japan. Int. J. Hematol. 2011; 94: 545– 80 Morimoto A, Ikushima S, Kinugawa N et al. Improved outcome
51. in the treatment of pediatric multifocal Langerhans cell
61 Grois N, Potschger U, Prosch H et al. Risk factors for diabetes histiocytosis: results from the Japan Langerhans Cell
insipidus in Langerhans cell histiocytosis. Pediatr. Blood Cancer Histiocytosis Study Group-96 protocol study. Cancer 2006; 107:
2006; 46: 228–33. 613–9.
62 Donadieu J, Rolon MA, Thomas C et al. Endocrine involvement 81 Morimoto A, Shioda Y, Imamura T et al. Intensified and pro-
in pediatric-onset Langerhans’ cell histiocytosis: a population- longed therapy improved the outcome in multi-system
based study. J. Pediatr. 2004; 144: 344–50. Langerhans cell histiocytosis. Pediatr. Blood Cancer 2013; 60:
63 Grois N, Prayer D, Prosch H, Minkov M, Pötschger U, Gadner H. E80. [abstract].
Course and clinical impact of magnetic resonance imaging find- 82 Minkov M, Grois N, Heitger A, Pötschger U, Westermeier T,
ings in diabetes insipidus associated with Langerhans cell Gadner H. Response to initial treatment of multisystem
histiocytosis. Pediatr. Blood Cancer 2004; 43: 59–65. Langerhans cell histiocytosis: an important prognostic indicator.
64 Imashuku S, Kudo N, Kaneda S et al. Treatment of patients with Med. Pediatr. Oncol. 2002; 39: 581–5.
hypothalamic-pituitary lesions as adult-onset Langerhans cell 83 Kudo K, Morimoto A, Imamura T et al. Outcome of poor
histiocytosis. Int. J. Hematol. 2011; 94: 556–60. responders at 6 weeks of induction therapy: JLSG-02 protocol

© 2014 Japan Pediatric Society


Recent advances in LCH 461

study for multisystem Langerhans cell histiocytosis in Japan. reduced-intensity conditioning. Bone Marrow Transplant. 2005;
Pediatr. Blood Cancer 2011; 56: 695. [abstract]. 36: 215–25.
84 Vassallo R, Ryu JH, Schroeder DR, Decker PA, Limper AH. 97 Morimoto A, Shioda Y, Imamura T et al. Nationwide survey of
Clinical outcomes of pulmonary Langerhans’-cell histiocytosis in bisphosphonate therapy for children with reactivated Langerhans
adults. N. Engl. J. Med. 2002; 346: 484–90. cell histiocytosis in Japan. Pediatr. Blood Cancer 2011; 56:
85 Girschikofsky M, Arico M, Castillo D et al. Management of adult 110–5.
patients with Langerhans cell histiocytosis: recommendations 98 Weitzman S, Braier J, Donadieu J et al. 2’-Chlorodeoxyadenosine
from an expert panel on behalf of Euro-Histio-Net. Orphanet J. (2-CdA) as salvage therapy for Langerhans cell histiocytosis
Rare Dis. 2013; 8: 72. (LCH). Results of the LCH-S-98 protocol of the Histiocyte
86 Yuasa M, Fujiwara S, Oh I et al. Rapidly progressing fatal adult Society. Pediatr. Blood Cancer 2009; 53: 1271–6.
multi-organ Langerhans cell histiocytosis complicated with fatty 99 Imamura T, Sato T, Shioda Y et al. Outcome of pediatric patients
liver disease. J. Clin. Exp. Hematop. 2012; 52: 121–6. with Langerhans cell histiocytosis treated with 2 chlorodeo-
87 Saven A, Burian C. Cladribine activity in adult Langerhans-cell xyadenosine: a nationwide survey in Japan. Int. J. Hematol. 2010;
histiocytosis. Blood 1999; 93: 4125–30. 91: 646–51.
88 Derenzini E, Fina MP, Stefoni V et al. MACOP-B regimen in the 100 Yamada K, Yasui M, Sawada A, Inoue M, Nakayama M, Kawa K.
treatment of adult Langerhans cell histiocytosis: experience on Severe persistent bone marrow failure following therapy with
seven patients. Ann. Oncol. 2010; 21: 1173–8. 2-chlorodeoxyadenosine for relapsing juvenile xantho-
89 Cantu MA, Lupo PJ, Bilgi M, Hicks MJ, Allen CE, McClain KL. granuloma of the brain. Pediatr. Blood Cancer 2012; 58: 300–
Optimal therapy for adults with Langerhans cell histiocytosis 2.
bone lesions. PLoS ONE 2012; 7: e43257. 101 Suzuki D, Kobayashi R, Sano H et al. Spontaneous remission of
90 Morimoto A, Shimazaki C, Takahashi S et al. Therapeutic 2-chlorodeoxyadenosine (2-CdA)-related secondary myelo-
outcome of multifocal Langerhans cell histiocytosis in adults dysplastic syndrome in a patient with refractory Langerhans cell
treated with the Special C regimen formulated by the Japan LCH histiocytosis. Int. J. Hematol. 2013; 97: 782–5.
Study Group. Int. J. Hematol. 2013; 97: 103–8. 102 Braier J, Ciocca M, Latella A, de Davila MG, Drajer M,
91 Bernard F, Thomas C, Bertrand Y et al. Multi-centre pilot study Imventarza O. Cholestasis, sclerosing cholangitis, and liver trans-
of 2-chlorodeoxyadenosine and cytosine arabinoside combined plantation in Langerhans cell histiocytosis. Med. Pediatr. Oncol.
chemotherapy in refractory Langerhans cell histiocytosis with 2002; 38: 178–82.
haematological dysfunction. Eur. J. Cancer 2005; 41: 2682–9. 103 Dauriat G, Mal H, Thabut G et al. Lung transplantation for pul-
92 Apollonsky N, Lipton JM. Treatment of refractory Langerhans monary Langerhans’ cell histiocytosis: a multicenter analysis.
cell histiocytosis (LCH) with a combination of 2- Transplantation 2006; 81: 746–50.
chlorodeoxyadenosine and cytosine arabinoside. J. Pediatr. 104 Minkov M, Steiner M, Pötschger U et al. Reactivations in multi-
Hematol. Oncol 2009; 31: 53–6. system Langerhans cell histiocytosis: data of the international
93 Simko SJ, Tran HD, Jones J et al. Clofarabine salvage therapy in LCH registry. J. Pediatr. 2008; 153: 700–5.
refractory multifocal histiocytic disorders, including Langerhans 105 Morimoto A, Kobayashi R, Maeda M, Asami K, Bessho F,
cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman Imashuku S. Impact of reactivation on the sequelae of multi-
disease. Pediatr. Blood Cancer 2014; 61: 479–87. system Langerhans cell histiocytosis patients. Pediatr. Blood
94 Haroche J, Cohen-Aubart F, Emile JF et al. Dramatic efficacy of Cancer 2008; 50: 931–2.
vemurafenib in both multisystemic and refractory Erdheim- 106 Haupt R, Nanduri V, Calevo MG et al. Permanent consequences
Chester disease and Langerhans cell histiocytosis harboring the in Langerhans cell histiocytosis patients: a pilot study from the
BRAF V600E mutation. Blood 2013; 121: 1495–500. Histiocyte Society-Late Effects Study Group. Pediatr. Blood
95 Kudo K, Ohga S, Morimoto A et al. Improved outcome of refrac- Cancer 2004; 42: 438–44.
tory Langerhans cell histiocytosis in children with hematopoietic 107 Urayama KY, Chokkalingam AP, Manabe A, Mizutani S. Current
stem cell transplantation in Japan. Bone Marrow Transplant. evidence for an inherited genetic basis of childhood acute
2010; 45: 901–6. lymphoblastic leukemia. Int. J. Hematol. 2013; 97: 3–19.
96 Steiner M, Matthes-Martin S, Attarbaschi A et al. Improved 108 Imadome K, Yajima M, Arai A et al. Novel mouse xenograft
outcome of treatment-resistant high-risk Langerhans cell models reveal a critical role of CD4+ T cells in the proliferation of
histiocytosis after allogeneic stem cell transplantation with EBV-infected T and NK cells. PLoS Pathog. 2011; 7: e1002326.

© 2014 Japan Pediatric Society

You might also like