Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jul 15.
Published in final edited form as: Cancer. 2024 Apr 30;130(14):2416–2439. doi: 10.1002/cncr.35301

Langerhans Cell Histiocytosis: NACHO Update on Progress, Chaos and Opportunity on the Path to Rational Cures

Kevin Bielamowicz 1,*, Peter Dimitrion 2,*, Oussama Abla 3, Simon Bomken 4, Patrick Campbell 5, Matthew Collin 6, Barbara Degar 7, Eli Diamond 8, Olive S Eckstein 9, Nader El-Mallawany 9, Mark Fluchel 10, Gaurav Goyal 11, Michael M Henry 12, Michelle Hermiston 13, Michael Hogarty 14, Michael Jeng 15, Rima Jubran 16, Joseph Lubega 9, Ashish Kumar 17, Stephan Ladisch 18, Kenneth L McClain 9, Miriam Merad 19, Qing-Sheng Mi 2, D Williams Parsons 9, Erin Peckham-Gregory 9, Jennifer Picarsic 20, Zachary D Prudowsky 9, Barrett J Rollins 21, Peter H Shaw 22, Birte Wistinghausen 18, Carlos Rodriguez-Galindo 23, Carl E Allen 9, on behalf of the North American Consortium for Histiocytosis
PMCID: PMC11214602  NIHMSID: NIHMS1983364  PMID: 38687639

Abstract

Langerhans cell histiocytosis (LCH) is a myeloid neoplastic disorder characterized by lesions with CD1a+/Langerin (CD207)+ histiocytes and inflammatory infiltrate that can cause local tissue damage and systemic inflammation. Clinical presentations range from single lesions with minimal impact to life-threatening disseminated disease. Therapy for systemic LCH has been established through serial trials empirically testing different chemotherapy agents and duration of therapy. However, fewer than 50% of patients with disseminated disease are cured with the current standard of care vinblastine/prednisone/(mercaptopurine), and treatment failure is associated with long-term morbidity including risk of LCH-associated neurodegeneration. Historically, the nature of LCH, whether a reactive versus neoplastic/malignant condition, was uncertain. Over the past 15 years, seminal discoveries have broadly defined LCH pathogenesis, specifically activating MAPK pathway mutations (most frequently BRAFV600E) in myeloid precursors drive lesion formation. LCH therefore is a clonal neoplastic disorder, though secondary inflammatory features contribute to disease. These paradigm-changing insights offer promise of rationale cures for patients based on individual mutations, clonal “reservoirs” and extent of disease. However, the pace of clinical trial development lags the kinetics of translational discovery. In this review, we discuss the current understanding of LCH biology, clinical characteristics, therapeutic strategies, and opportunities to improve outcomes for every patient through coordinated agent prioritization and clinical trial efforts.

Keywords: Langerhans cell histiocytosis, MAPK pathway, myeloid neoplasia, clinical trials

PRECIS

Langerhans cell histiocytosis is an inflammatory myeloid neoplastic disorder driven by activating somatic mutations in MAPK pathway genes. Rapid advances in mechanistic understanding of LCH now offer opportunities to improve outcomes for patients.

Introduction

This manuscript was organized through collaboration between the North American Consortium for Histiocytosis members and partners in the United Kingdom to review the current biological understanding of Langerhans cell histiocytosis (LCH) (progress), data to guide current therapeutic approaches in the context of rapidly-changing biological paradigms and development of novel agents (chaos), and strategies to improve therapy for patients (opportunity).

Background

The epidermal Langerhans cell was first described in 1868 by the Berlin medical student Paul Langerhans as a neuronal cell because of its distinct dendrite-like processes.1 In a subsequent article, “Corrections”, he withdrew his initial assertion and speculated that this cell might be of hematopoietic origin.2 Between 1893 and 1919, Hand, Schüller, and Christian separately published the first clinical descriptions of Langerhans cell histiocytosis (LCH) of patients with exophthalmos, lytic bone lesions and diabetes insipidus.35 In 1924, Letterer reported an infant with extensive skin rash, pancytopenia, hepatosplenomegaly and tachypnea followed nine years later by Siwe describing a toddler with similar symptoms.6,7 Initially considered distinct entities, isolated bone lesions were called eosinophilic granulomas, multifocal bone disease with diabetes insipidus, Hand-Schüller-Christian Disease, and the most severe multi-system disorder, Letterer-Siwe Disease. In 1941, Farber noted these entities to have similar histologic features. In 1953, Lichtenstein concluded that these were different clinical manifestations of the same disease affecting the reticuloendothelial system and proposed the name “histiocytosis X” to reflect the unknown origin of the lesions cells, thus conceptualizing “histiocytoses”.8,9 However, electron microscopy revealed Birbeck granules in those cells, leading Nezelof to hypothesize a developmental link with the epidermal Langerhans cell in 1973.10 This led to the renaming of histiocytosis X to LCH.10

A long-standing debate as to whether LCH was a malignancy or an inflammatory disease continued even after the clonality of LCH was established in the 1990s11,12. Subsequently, BRAFV600E (or other MAPK activating mutations) in myeloid precursors were identified as pathogenic drivers, thus placing LCH in the category of myeloid neoplasms and conclusively demonstrating that LCH is an oncologic process.1315 This discovery contributed to the current understanding of LCH as both a malignancy AND an inflammatory disorder.16 While infection or immune dysregulation may not be drivers of disease, inflammation induced by MAPK-activated myeloid cells contributes to pathogenic features.

Diagnosis and Classification

Histology

Once the possibility of LCH is considered, its histopathologic diagnosis is straightforward in most cases. LCH lesions are characterized by CD1a+ Langerin (CD207+) dendritic cells (DC) with reniform nuclei in the setting of a mixed inflammatory infiltrate (Figure 1). Birbeck granules are characteristic of LCH dendritic cells, but electron microscopy is no longer an essential diagnostic tool in the presence of CD1a and CD207 (Langerin) expression which are surrogate diagnostic markers.17 Peculiarities of staining intensity of Langerin, a component of the Birbeck granule apparatus, can confound the diagnosis in some cases. Surface Langerin is expressed in normal, physiologic epidermal Langerhans cells. Therefore, in addition to Langerin expression, cell features (e.g., dendritic cell (DC) morphology) and pattern of distribution differentiate epidermal LC/DC from LCH cells in skin and draining lymph nodes (Figure 1)18. Additionally, Langerin expression is induced in precursors and not a fixed feature of LCH cells: Langerin is dim or absent in many cases of liver, bone marrow and CNS disease.14,19 Increasing use of pan-histiocytosis diagnostic markers is also important in identifying mixed histiocytic lesions (e.g. LCH with juvenile xanthrogranuloma (JXG), Rosai-Dorfman disease (RDD) and/or Erdheim-Chester disease (ECD)). Additionally, identification of molecular alterations (e.g. mutant specific BRAF VE1 stains, BRAFV600E qPCR, targeted next generation sequencing) support histiocytic neoplasm diagnosis and identify potential therapeutic targets.

Figure 1. LCH Histology.

Figure 1.

LCH lesions are characterized by “histiocytes” with reniform nuclei and surrounding inflammatory infiltrate. (A) H&E (80x), (B) CD1a immunohistochemistry (80x), (C) Electron microscopy demonstrating Birbeck granule, which is now replaced with Langerin (CD207) stain (not shown). LCH can be challenging to differentiate from reactive Langerhans cells/dendritic cells in skin and lymph nodes (LN). Skin with reactive CD1a+ dendritic cell hyperplasia in the dermis with perivascular histiocytes that are CD1a+ with spindled dendritic shape (D-E) are low to negative Langerin/CD207 staining (F) (10x). By comparison, (G-I) shows a case of skin with superficial dermal infiltrate of LCH with epidermotropism and surface ulceration. The CD1a (H) and Langerin (I) stains highlight the clustering of plump lesional histiocytes in the papillary dermis (10x). Activated DC drain into LN to present antigen to lymphocytes as demonstrated by hyperplasia of reactive dendritic/Langerhans cells in the paracortex ((J) H&E, (K) CD1a, (L) Langerin, 13x digital images). By comparison, nodal LCH is characterized by sinus expansion of LCH with variable spill over into the paracortex ((M) H&E, (N) CD1a, (O) Langerin, 10x). (Images courtesy of Jennifer Picarsic, MD, Cincinnati Children’s Hospital, Cincinnati, OH).

Classification

Histiocytic disorders have historically been classified according to terminal phenotype and similarity to physiologic cells of the mononuclear phagocyte system2022 (e.g. “Langerhans cell” histiocytosis because of overlapping features with epidermal LC). However, the past 15 years have seen notable advances in mechanistic understanding of LCH pathogenesis and phylogeny. Based on these developments, a revised reclassification strategy was proposed that includes not only cell phenotype, but also clinical patterns and molecular drivers.23 LCH is housed in the “L-group” that also includes ECD and disseminated JXG. However, the updated 5th edition World Health Organization (WHO) classification of mature lymphoid, histiocytic, and dendritic neoplasms separates ECD and LCH into two distinct entities based on integration of clinical, radiological, and histopathological diagnosis.24 (Table 1)

Table 1.

Classifications of Histiocytic Disorders.23,24

WHO 5th Edition Classification of Haematolymphoid Tumors: Dendritic cell and histiocytic neoplasms
Plasmacytoid dendritic cell neoplasms
  • Mature plasmacytoid dendritic cell proliferation associated with myeloid neoplasm

  • Blastic plasmacytoid dendritic cell neoplasm

Langerhans cell and other dendritic cell neoplasms
Langerhans cell neoplasms
  • Langerhans cell histiocytosis

  • Langerhans cell sarcoma

Other dendritic cell neoplasms
  • Indeterminate dendritic cell tumor

  • Interdigitating dendritic cell sarcoma

Histiocytic neoplasms
  • Juvenile xanthogranuloma

  • Erdheim-Chester disease

  • Rosai-Dorfman disease

  • ALK-positive histiocytosis

  • Histiocytic sarcoma

Histiocyte Society Revised Classification Proposal
L Group
  • Langerhans cell histiocytosis

  • Erdheim-Chester disease

  • Disseminated juvenile xanthogranuloma

  • ALK-positive histiocytosis

  • Indeterminate cell histiocytosis

  • Mixed LCH/ECD

C Group
  • Skin limited JXG

  • Other skin-limited xanthogranulomas and non-Langerhans histiocytoses

R Group
  • Familial Rosai-Dorfman disease (RDD)

  • Classical (nodal) RDD

  • Extranodal RDD

  • Neoplasia-associated RDD

  • Immune-associated RDD

M Group
  • Primary malignant histiocytosis

  • Secondary malignant histiocytosis (associated with other hematologic malignancy)

H Group
  • Primary hemophagocytic lymphohistiocytosis (HLH)

  • Secondary HLH

  • HLH of uncertain origin

Evolving Understanding of LCH Biology

Prior to 2010, clues regarding the origins and nature of LCH came primarily from histology. Characteristic but enigmatic features led to recurrent debate regarding LCH as a reactive inflammatory (e.g. immune infiltrate, rare mitoses, no known mutations, rare spontaneous regression) versus neoplastic disorder (e.g. persistence and progression, clonality, destructive tumors, response to chemotherapy, lack of associated infection).16 Discovery of recurrent BRAFV600E mutations13 followed by reports of near universal and mutually exclusive activating MAPK pathway mutations2527, and an otherwise quiet genome highlighted the importance of MAPK activation in this disease’s pathophysiology. Physiologic MAPK signaling enables cells to differentiate, proliferate and survive in response to extracellular environmental cues (Figure 2A).28 BRAFV600E mutations were discovered in CD34+ hematopoietic stem cells and in peripheral blood of all patients with high risk (HR) multisystem LCH, less frequently in patients with low risk (LR) single system disease, and very rarely in patients with single lesions. BRAFV600E activates MAPK signaling independent of upstream regulation by receptor tyrosine kinases (RTK) or Ras-family GTPases (Figure 2A).29

Figure 2. LCH Pathogenic Mechanisms.

Figure 2.

(A) Diagram of the MAPK signaling pathway. When growth factors/mitogens bind to receptor tyrosine kinases (RTKs), RTKs activate guanine exchange factors to exchange GDP for GTP on Ras, which activates Ras. Normally, GTP bound Ras is required for the downstream activation and homodimerization of Rafs (BRAF or ARAF) and further signaling components of MAPK. The BRAFV600E allows BRAF to function as a monomer independent of Ras activation, which leads to constitutive activation of MAPK signaling pathway. (B) Studies in patients with LCH have shown that acquisition of allelic MAPK-activating allelic variants in myeloid precursors leads to HR LCH due to pathological myeloid cells having access to the circulation, supported by the finding that HR-patients (but never LR-patients) harbor circulating mutation bearing myeloid cells. Circulating CD11a+ myeloid cells also contribute the LCH-ND. This risk-stratification can be modeled using genetic knock-in models where expression of BRAFV600E can be enforced in hematopoietic stem cells (SclCre BRAFV600Elox-stop-lox; BRAFV600Escl or Map17Cre BRAFV600Elox-stop-lox; BRAFV600EMap17) or the entire DC lineage (CD11cCre BRAFV600Elox-stop-lox; BRAFV600ECD11c), which lead to lethal LCH-like disease. In comparison, enforced expression of BRAFV600E in tissue limited DCs (CD207Cre BRAFV600Elox-stop-lox; BRAFV600CD207) form lesions, but do not die from LCH-related causes. (C) Enforced expression of BRAFV600E in mouse models have found that enhanced myeloid differentiation, so called “myeloid skewing”; activation and exhaustion of T cells, tissue entrenchment by decreased CCR7 expression, and resistance to apoptosis promote the accumulation of pathological DCs in nearly any organ. The latter is part of the BRAFV600E-induced SASP, which is central to LCH pathophysiology.

Enforced expression of BRAFV600E in differentiated CD207+ cells in mice led to development of some lesions, but overall was a mild phenotype. By comparison, expression of BRAFV600E+ in myeloid cells and precursors (CD11ccre, Sclcre and MAP17cre) in mice drove an aggressive HR LCH-like phenotype where mice survived only a few months.14,30,31 Together, these data from patients and mice support a model in which MAPK activation in myeloid precursors drives disseminated LCH, while less extensive disease is associated with the same MAPK pathway alterations in more differentiated myeloid cells (“the misguided myeloid DC model of LCH pathogenesis”) (Figure 2B).32

MAPK activation in myeloid precursors enhances differentiation toward the myeloid lineage (e.g. “myeloid skewing”. MAPK-activated myeloid DC lack CCR7 expression, which is required for physiologic migration of DC toward lymph nodes. Further, MAPK activation in lesion DC induces apoptosis resistance via upregulation of Bcl-xL, activation and exhaustion of infiltrating T cells via checkpoint inhibition, and induction of an oncogene-induced senescence program.30,33 Together, enhanced differentiation, entrenchment, resistance to apoptosis, and the ability to orchestrate tissue immune responses lead to accumulation of pathological DCs and formation, persistence and progression of LCH lesions (Figure 2C).

Beyond somatic mutations, a case-parent trio study using genome-wide association methodology identified inherited risk allele in SMAD6.34 Non-random racial and ethnic distribution (e.g. increased risk in AmerIndian/Hispanic, decreased risk in African ancestry) suggests germline variants may influence LCH risk. LCH does not typically present in families, with the exception of monozygotic twins who share hematopoietic origins. Pediatric LCH arises in approximately 5 children per million, and is estimated to arise in 1 adult per million.35,36

Clinical Manifestations of LCH in Children

LCH can affect any organ in the body and is typically categorized as single lesion, single-system unifocal, single-system multifocal or multi-system based on extent of disease. (Figure 3). Common sites of disease include skin, bone, lung, liver, and the pituitary gland. Multisystem LCH is further stratified into low- or high-risk groups based on the organs affected and corresponding mortality risks (also referred to as risk organ positive or negative (RO+/RO-); also called “high risk” (HR) and “low risk” (LR)). Specific “risk organ” systems (e.g., liver, spleen, bone marrow) were so classified based on their association with higher mortality risks in the LCH-II trial.37 Within these risk groups, clinicians have recognized classical patterns of presentation. For example, HR disseminated LCH is most common in infants, multifocal disease without risk organ involvement is more common in preschoolers, and more than 50% of patients with an isolated bone lesion are diagnosed after the age of 5 years. Additionally, “CNS risk” lesions, defined as those involving facial bones and the skull base, are associated with development of LCH-associated pituitary dysfunction and neurodegeneration.38 The relatively worse prognosis with liver, spleen and bone marrow disease applies to adults as well as children.39 The diagnosis of LCH can be invoked frequently at residency problem conference “morning report” diagnostic dilemma seminars as the vast spectrum of clinical features can mimic more common conditions (Table 1, Figure 3).

Figure 3. Clinical Features of LCH.

Figure 3.

Positron-emission tomographic (PET) images show a single bone lesion involving the humerus (Panel A, arrow); low-risk lesions involving the orbit, lymph nodes, bone (multifocal lesion), and thymus (Panel B); and high-risk lesions involving the liver, spleen, and bone marrow (Panel C). Other classic presentations include a lytic bone lesion (Panel D, arrow), cystic lung lesions (Panel E), and various skin lesions (Panels F through I). Examples of LCH lesions involving the skull and brain include multifocal skull lesions (Panel J, arrow), an orbital lesion (Panel K, arrow), a pituitary lesion (Panel L, arrow), and LCH-associated neurodegeneration (Panel M, arrow). (Figure from Allen CE et al. N Engl J Med 2018;379:856–868).

Skin, either isolated or with additional affected systems, is a common site of disease in all ages. The diagnosis of skin-limited LCH should not be presumed from a skin biopsy alone as 40% of children with presumptive skin-limited disease were diagnosed with multi-system disease on further investigation.40 LCH limited to the skin has a very favorable outcome. In a retrospective study, patients with skin-limited disease had a 3-year progression-free survival (PFS) of 89% compared to 44% for patients with skin and multi-system involvement.40 Notably, peripheral blood BRAFV600E+ cells are typically undetectable in patients with truly isolated skin LCH, but are found in patients with skin LCH as part of systemic disease.40 This pattern, along with potential for isolated skin LCH to spontaneously resolve, support a concept of isolated skin LCH arising from tissue-restricted erythromyeloid precursors (tissue resident macrophages/epidermal LCs) while systemic disease arises from hematopoietic precursors. (Figure 3) Skin lesions vary in clinical presentation from dermatitis, vesicular eruptions, ulcerative lesions, or petechial rashes. Although any area of skin may be involved, LCH most commonly involves the scalp, axilla, and perineum. In infants, it commonly resembles seborrheic dermatitis and can also be mistaken for persistent cradle cap. A self-limited rash in infants known as Hashimoto-Pritzker is characterized by darker papular lesions. In older children, skin lesions more often develop in skin folds may be misdiagnosed as fungal infection.41 Nail involvement is rare, presenting as discoloration, hardening of nail beds or loss of nail tissue.42 Persistent otorrhea suggests otologic involvement, which can mimic infection. Oral manifestations may include ulcers and gingival hypertrophy with the potential for underlying bone involvement and tooth loss.43

Bone disease may present either as uni- or multifocal disease and can be associated with soft tissue masses. While LCH can occur in any bone, children commonly present with lytic skull lesions manifesting as pain and/or swelling.44 Vertebral bone involvement can cause vertebral collapse or vertebra plana.45 Bone lesions affecting the skull base, including orbital, mastoid and temporal bones, increase the risk of CNS involvement.38

Lung involvement can occur in any age. In contrast to children, lung LCH in adults can arise in smokers, presumably precipitated by reaction to smoke-related inflammation, and can resolve with smoking cessation alone.7 Lung involvement is found in 25% of children with multisystem disease. While pulmonary LCH can cause dramatic lung injury in some cases, lung disease is not considered an independent prognostic factor for moratlity.46 Lung involvement may be asymptomatic and diagnosed on imaging as symmetrical areas of nodular fibrosis and bullae formation affecting the upper and middle lobes.47 Rarely, patients complain of cough, shortness of breath or develop spontaneous pneumothoraces. In more severe cases, advanced lung damage can lead to hypoxia and restrictive lung disease with irreversible fibrosis in some cases.48

Lymph nodes in the cervical chain are the most commonly affected and present as enlarged, matted, soft or hard nodes. Mediastinal involvement with thymic or lymph node infiltration is rare.49

Central nervous system involvement can present with lesions in the hypothalamic-pituitary region, dural- based masses, infiltration in the choroid plexus or changes in white mater in the basal ganglia and cerebellum.50 Involvement of the hypothalamic-pituitary region can be isolated or a component of multisystem disease. The posterior pituitary is affected most frequently and can lead to diabetes insipidus (DI). Approximately 50–80% of patients with pituitary involvement and DI will develop other manifestations of LCH5153 Patients with multisystem disease and craniofacial bone involvement at the time of diagnosis are at increased risk for developing DI (Relative risk 4.6).54 On magnetic resonance imaging (MRI), loss of the pituitary bright spot, often with a nodular mass or thickening of the pituitary stalk is noted.55 Panhypopituitarism can also develop with growth hormone deficiency occurring in 25% of patients with DI.56 Dural-based mass lesions may be noted incidentally or present with symptoms of space-occupying lesions.55

Neurodegenerative CNS disease (LCH-ND) develops in approximately 10% of patients with LCH. Patients with pituitary involvement and/or craniofacial bone lesions are at increased risk for LCH-ND, and >90% of patients with LCH-ND have BRAFV600E-driven disease5759 This complication typically involves the cerebellum, basal ganglia and pons. “LCH-associated abnormal CNS imaging” (LACI) describes LCH-ND with imaging changes only whereas “LCH-associated abnormal CNS symptoms” (LACS) describes LCH-ND with clinical symptoms. Patients with LACS have a variable clinical course, including dysmetria, tremor, ataxia, dysarthria, behavioral disturbances, cognitive disorders and/or psychosis which can occur many years after the initial diagnosis of systemic LCH. Imaging on MRI shows hyperintensity on T1 weighted images in the dentate nucleus with hyper- or hypointensity on T2 weighted images. MRI changes can precede neurologic symptoms, or in some cases can present in patients who remain asymptomatic with “LCH-associated abnormal CNS imaging” (LACI).38 Some patients develop extension into the white matter of the cerebellum and frequently associated hyperintensity in the basal ganglia on T1 weighted images. Involvement of the pons is associated with particularly severe neurologic impairment.50,60,61 LCH-ND has historically been considered an immune reaction or paraneoplastic phenomenon in patients with a history of LCH. However, recent studies identified peripheral blood BRAFV600E+ myeloid cells in patients with LCH-ND in the absence of systemic disease, and brain biopsies demonstrated BRAFV600E+ monocyte/macrophage cells enriched at sites of LCH-ND.58 Further, in a recent mouse model, senescent BRAFV600E+ CD11A+ monocyte-derived meningeal macrophages were able to break through the blood-brain barrier and drive neurodegeneration in patterns similar to human LCH.31 These data together support a hematopoietic origin for clonal myeloid cells with the potential for peripheral precursors to migrate to the brain and promote chronic neurodegeneration in LCH-ND. Notably, BRAFV600E+ T cells are observed in peripheral blood of some patients who develop LCH-ND. It is not known if these T cells are vestigial long-surviving clones or if they might play a role in LCH-ND pathogenesis.62

Bone marrow involvement in LCH presents as cytopenias associated with multisystem disease and carries an inferior prognosis.6365 Anemia is the most common cytopenia, followed by thrombocytopenia. Bone marrow findings include hemophagocytosis or increased macrophages, which may be reactive or harboring BRAFV600E or other MAPK gene mutations. Quantitative PCR and/or immunohistochemistry with BRAF VE1 (which stains for BRAF-V600E mutant protein) demonstrate mutated cells in the bone marrow with much higher frequency than conventional morphology or CD1a/Langerin immunohistochemistry.14,66

Liver involvement can be seen in infants and toddlers, and it is commonly associated with multisystem disease. Transcutaneous biopsy of the liver may be non-diagnostic of CD1a+/Langerin+ cells given the large duct involvement, but typically will show biliary obstructive features. LCH involvement tends to occur around bile ducts and cause ductal sclerosis. Signs of liver dysfunction include direct hyperbilirubinemia, cholestasis, hypoalbuminemia, coagulopathy and ascites.67,68 On radiographic imaging, involvement may be nodular or diffusely infiltrative. Sclerosing cholangitis is a rare sequela that may not respond to therapy in cases of long-standing liver damage (similar to entrenched LCH-ND). Liver transplant has been used with varying success.69,70 Involvement of the spleen usually occurs along with the liver. Hypersplenism can cause cytopenias.

Intestinal involvement presents with bloody diarrhea, failure to thrive and malabsorption. Histological diagnosis can be difficult due to patchy involvement. However, demonstration of mutant histiocytic cell involvement, either bys ensitive molecular PCR or BRAF VE1 staining in known BRAFV600E LCH cases, may be used to infer intestinal disease.7173

Other organs less commonly involved in LCH include the thyroid, which can present with thyromegaly and/or hypothyroidism74 and eyes, which can result in visual impairment.75

LCH in Adults

Adults can develop LCH de novo or carry unresolved LCH from childhood. Clinical presentation and diagnostic approaches are similar between adults and children with some caveats.39 In general, LCH arises in children due to accidents of hematopoietic development and in adults due to hematopoietic decline. Adults typically have more somatic mutations than children, though still with a relatively low burden.76 LCH can arise in some cases along with evolving hematopoietic stem cell mutations during clonal hematopoiesis.77 LCH arising as part of a “mixed histiocytic” disorder (e.g. ECD and LCH features in the same lesion or ECD and LCH histology in distinct lesions) occurs in approximately 5% of cases.78 Isolated pulmonary LCH is a unique adult feature, almost exclusively in smokers. While data is lacking to describe quality of life and patient-perceived symptoms for adults with LCH, a patient-reported outcome study identified significant pain and fatigue in the majority of patients with ECD.79 Patients with long-standing systemic disease and/or concerning neurologic symptoms and/or peripheral blood BRAFV600E+ cells may warrant MRI evaluation for LCH-ND.

Staging

A comprehensive evaluation for each patient is important to determine the extent of disease and organ systems involved. This is done through history, physical examination, laboratory studies and imaging. PET-CT is highly sensitive for systemic lesions.80 PET-MR is also feasible if available and if the patient is able to tolerate the increased acquisition time.81 Skeletal survey and abdominal ultrasound may be used when PET-CT is not available and in infants for whom PET-CT may not be practical. MRI is preferred for evaluation of brain and spinal cord lesions, and T2-weighted brain MRI is optimal for LCH-ND.38 Risks and benefits of any imaging study needs to take into account risks (e.g. radiation exposure, need for prolonged anesthesia) along with diagnostic sensitivity. Bone marrow aspirate and biopsy may be informative for patients with cytopenias at presentation and may be considered for any patient under 2 years of age with multisystem disease. Bone marrow aspirate and peripheral blood BRAFV600E qPCR or ddPCR can identify LCH precursors in patients with BRAFV600E+ lesions that may become useful for risk stratification and evaluation of minimal residual disease which is discussed below. For adults, bone marrow studies can also identify potential co-incident development of clonal hematopoiesis/myelodysplasia.14,8284

Current Therapeutic Strategies

Identifying optimal therapy for LCH has been confounded, until recently, by limited mechanistic understanding of the disease. Uncertainty of LCH as a reactive immune disorder versus neoplastic disorder (e.g., cancer, or cancer family) has led to questions regarding the optimal therapeutic approach (e.g., immune suppression vs chemotherapy), treatment goal (e.g., management of flare vs cure), and response nomenclature (e.g. “reactivation” vs “relapse”).

Front-Line – Limited Disease

Single lesions.

There have been few clinical trials to guide therapy for single LCH lesions, so practice is based largely on consensus and expert opinion.85 Biopsy/curettage with or without local steroid injection may be sufficient therapy for single bone lesions.86 Due to association of lesions in skull base and facial bones with risk of LCH-ND, systemic chemotherapy is recommended, even for single bone lesions in those locations. Similarly, systemic therapy is typically recommended for LCH involving the pituitary to decrease risks of developing DI and LCH-ND.38

Bone lesions.

Patients with single site and multi-focal bone LCH should undergo a full staging work-up. A recent report recommended observation only in presumed isolated non-CNS risk skull lesions of the calvarium with the assumption that these will spontaneously resolve.87 However, biopsy can diagnose other conditions (e.g. infection, vascular malformation, other malignancy) that may require urgent alternative therapy, and comprehensive imaging is important to confirm that a lesion is in fact “isolated”. The risks of biopsy are minimal, and the procedure may offer therapeutic benefit in true isolated LCH lesions. On the other side of the surgical spectrum, gross total resections are also not warranted because most single lesions with intact margins typically resolve and remodel following curettage/local steroid injection or systemic chemotherapy. Low-dose radiation therapy, which can also be effective in emergency situations for resolution of individual lesions, is mostly restricted to adult patients. Multifocal bone disease is a common presentation of LCH, and we typically consider single-system, multi-focal bone LCH to represent systemic disease.

Skin lesions.

As discussed above, skin LCH can represent the tip of a multi-system LCH “iceberg” or truly be skin-limited.40 Most cases of skin-limited disease arise in infants and resolve spontaneously. Topical steroids, methotrexate and/or hydroxyurea may improve symptoms. Conventional systemic LCH therapy can be used in rare refractory cases (although these cases likely represent disseminated disease).

Front-Line – Systemic Disease

The Histiocyte Society Trials (LCH I-III) built on the DAL-HX studies (collaborations between European institutions) to test risk-adapted approaches through randomized trials (Table 3)88,89. We consider the current standard of care for front-line therapy for pediatric patients with multifocal disease or single lesion in risk organ or CNS-risk site to be 1 year of vinblastine/prednisone (with 6-mercaptopurine (6MP) for HR LCH). This practice is based on the Histiocyte Society LCH-III trial.90 A major conclusion from the LCH-III trial is that extended therapy (e.g. 12 vs 6 months) was associated with decreased risk of relapse in patients with multisystem LR LCH. We extrapolate this result to suggest that increased duration of therapy may be beneficial in patients with systemic LCH in general (e.g. multifocal single system and CNS-risk in addition to multisystem LCH). Verification of this hypothesis is being tested in LCH-IV.

Table 3. Front-line pediatric LCH trials.

PFS-progression-free survival; OS-overall survival; EFS-event-free survival; HR-high risk (risk-organ positive); LR-low risk (risk-organ negative); MS-multisystem; SS MF-single system multifocal bone; 6MP-6-mercaptopurine; vin-vinblastine; P-prednisone/prednisolone; Etop-etoposide; mo-month

Study Time Period Groups (number) Therapies Responses OS Conclusions
DAL-HX83
DAL-HX90 88
1983–1991
  1. LR (Multifocal bone), n=28

  2. LR (Bone + soft tissue), n=57

  3. HR, n=21

  • Induction – vinblastine, prednisone, etoposide (all)

  • Continuation
    1. vinblastine, prednisone, 6MP
    2. vinblastine, prednisone, etoposide
    3. vinblastine, prednisone, 6MP, etoposide, methotrexate
  • 1 year

Disease resolution on study
  • Total-86%
    • A-89%
    • B-91%
    • C-67%
8 years
  • Total-81%
    • A-100%
    • B-96%
    • C-62%
  • Extent of disease at presentation is an important prognostic factor.

  • Response to induction is an important prognostic factor

LCH-I 89 1991–1995 LR+HR, n=143
  • Single high dose methylprednisolone pulse for both arms

  • Randomization:
    • Vinblastine
    • Etoposide
  • 24 weeks

Response at last eval (median 4 year 11 month)
  • Vinblastine – 58%

  • Etoposide – 65%

3 year “reactivation after NAD”
  • Vinblastine – 61%

  • Etoposide – 55%

3 years
  • Vinblastine- 76%

  • Etoposide- 83%

  • No statistical difference in survival probability between groups.

  • Worse 6 week response (50 vs 80%) and more relapse (50 vs 30%) than more intense and longer DAL-HX83.

  • Lack of response by 6 weeks recognized as a poor risk factor.

LCH-II 37 1996–2001 LR+HR, n=193
  • Randomization
    • Vinblastine, prednisone, 6MP
    • Vinblastine, prednisone, 6MP, etoposide
  • 24 weeks

1 year “NAD”
  • Vin/P/6MP – 49%

  • Vin/P/6MP/Etop– 62%

3 year “reactivation after NAD”
  • Vin/P/6MP – 46%

  • Vin/P/6MP/Etop– 46%

5 years
  • Vin/P/6MP – 74%
    • HR-64%
    • LR-100%
  • Vin/P/6MP/Etop – 41%
    • HR-73%
    • LR-100%
  • Therapy intensification associated with improved response rate and decreased mortality relative to LCH-I

  • 6 months of therapy appeared inferior to historic 12 month DAL-HX studies

  • Supported “high risk/RO+” and “low risk/RO-” designations

JLSG-96 91 1996–2001 SS MF and MS, n = 91
  • Prospective, non-randomized
    • Cytarabine, Vincristine, Prednisone, methotrexate
    • Intensification Regimen B if poor response
  • 24 weeks

5 year EFS
  • SS MF – 68.5%

  • MS – 28.8%

5 years
  • SS MF – 100%

  • MS – 94.4%

  • Response rates and OS similar to contemporary front-line regimen

  • Higher relapse rates may be due to short duration of treatment

JLSG-02 122,123 2002–2009 SS MF and MS, n = 91
  • Prospective, non-randomized
    • Cytarabine, Vincristine, Prednisone, methotrexate
    • Intensification regimen B if poor response
  • 48 weeks

5 year EFS
  • SS MF – 66.7%

  • MS – 46.2%

5 years
  • SS MF – 100%

  • MS – 91.7 %

  • Longer duration of therapy associated with improved EFS in MS, but not in SS MF pediatric LCH patients

LCH-III 90 2001–2008
  1. HR, n=235

  2. MS LR, n=187

  • HR Randomization
    • Vinblastine, prednisone, 6MP,
    • Vinblastine, prednisone, 6MP, methotrexate
    • 1 year
  • LR Randomization
    • Vinblastine, prednisone × 6 months
    • Vinblastine, prednisone × 1 year
1 year “NAD”
  • HR-no MTX – 48%

  • HR-MTX – 48%

5 year reactivation after “NAD”
  • LR-6 mo arm – 54% at 5 yr

  • LR-12 mo arm – 37% at 5 yr

5 years
  • HR-no MTX – 88%

  • HR-MTX – 82%

  • LR-6 mo – 100%

  • LR-12 mo – 99%

  • Addition of methotrexate did not significantly impact outcomes for HR LCH

  • Therapy prolongation decreased reactivation rates.

The majority of patients with multi-system LCH treated with one year of vinblastine/prednisone/6MP (according to LCH-III) will require salvage therapy.90 This has prompted the question of whether vinblastine/prednisone/6MP should be considered the standard of care. However, prospective clinical trial data supporting alternative regimens is currently lacking. Additionally, likely due to its roots as a potential “inflammatory disorder”, initial failure has been tolerated for LCH more than is typical for front-line cancer therapy. The LCH-III-based therapy has proven to be relatively well-tolerated in children with low risk of long-term sequelae.90

The Histiocyte Society LCH-IV trial tests hypotheses generated from prior European and Histiocyte Society trials, studies conducted through groups such as the Japan Langerhans Cell Histiocytosis Study Group (JLSG),91 as well as observations from retrospective series.92,93 The LCH-IV front-line stratum is now evaluating the impact of 24 vs 12 months of vinblastine/prednisone and inclusion or exclusion of 6MP in pediatric LCH with prospective double-randomization (NCT02205762). Another multi-site study, LCH-REASON (NCT02670707) is evaluating vinblastine/prednisone/6MP versus cytarabine monotherapy. The recent shift in practice at some institutions to treat patients exclusively with MAPK inhibition is discussed below.

Chemotherapy Salvage/Hematopoietic Stem Cell Transplant

Data to guide treatment after front-line failure is limited (Table 4). Case series and early-phase studies demonstrate high response rates to nucleoside analogs (e.g., cladribine, cytarabine, clofarabine), but durability of response is related to dose, duration and intensity. For example, only 4% of patients treated with low-dose cladribine (5 mg/m2/day × 5 days/cycle) on the LCH-S-98 trial were cured after 6 months.94 By comparison, in the LCH-S-2005 trial, the 5-year overall survival and progression-free survival were 85% using high-dose cladribine (9 mg/m2/day) with cytarabine (1 g/m2/day). However, this regimen was associated with significant toxicity (8% treatment-related mortality).95 Case series support potential therapeutic potential of intermediate dose of single agent cytarabine96 or clofarabine9799 that can typically be tolerated in outpatient settings. Oral hydroxyurea, a standard of care for myeloproliferative neoplastic disorders in the pre-tyrosine kinase inhibitor era, may also be effective for patients with LCH.100 The relative importance and risks of intensity versus duration in LCH therapies is a fundamental question that remains unanswered. Prospective trials are needed to determine the “just right” agent, dose and schedule for specific risk groups.

Table 4. Salvage chemotherapy trials for pediatric LCH.

HR-high risk (risk-organ-positive); LR-low risk (risk-organ-negative); NAD-no active disease; ORR-overall response rate ((NAD + active disease-better); or (CR + PR)); OS-overall survival; PFS-progression-free survival; CR-complete response; PR-partial response; SD-stable disease; PD-progressive disease; TRM-treatment-related mortality

Study Design Subjects Therapies Response Timepoint Outcomes Conclusions
LCH-S-98 94 Phase 2 Total-83
  • 46 HR

  • 37 LR

Cladribine (5 mg/m2/day × 5 days)
  • 21 day cycles

  • 2–6 cycles

6 month
2 year
NAD – 4%
  • LR – 10%

  • HR – 0%

ORR – 40%
  • LR- 62%

  • HR-23%

OS – 68%
  • LR-97%

  • HR-48%

  • Lower dose cladribine achieved responses in many patients with relapsed/refractory LCH, but achieved durable complete responses in very few.

  • An adult LCH series reported 67% ORR for cladribine salvage.124

LCH-S-2005 95 Phase 2 Total – 27 HR Cladribine (9 mg/m2/day) + cytarabine (1 g/m2/day) × 5 days
  • 28 day cycles

128 day (median)
2 month
5 year
NAD – 85%
ORR – 92%
OS – 85%
  • Very high dose nucleoside analog combination therapy yielded relatively high response rates, and also significant rates of treatment-related mortality (7.4%).

  • Most patients successfully completing therapy were “cured” of LCH.

Cytarabine monotherapy 96 Retrospective Total – 22
  • 6 HR

  • 16 LR

Cytarabine (100–170 mg/m2/day) × 5 days every 4 weeks for 6–12 months. 3 month
3 year
3 year
ORR – 59%
PFS-41%
OS-100%
  • Intermediate (typically outpatient) dose cytarabine monotherapy can elicit durable responses in patients with relapsed and refractory LCH.

Clofarabine monotherapy 99 Retrospective Total – 11
  • 3 HR

  • 8 LR

Clofarabine (25 mg/m2/day) × 5 days every 4 weeks for 6–12 months. 2 month
1 year
1 year
ORR – 73%
PFS-76%
91%
  • Intermediate (typically outpatient) dose clofarabine monotherapy can elicit durable responses in patients with relapsed and refractory LCH.

  • Other series with similar outcomes.97,98

Hydroxyurea oral therapy 100 Retrospective Total – 15
  • Adult – 12

  • Pediatric – 3

  • HR-2

  • LR-13

Hydroxyurea (500 mg twice daily or 20 mg/kg)
  • 5 also treated with oral methotrexate

1–24 months ORR – 80%
  • CR-8

  • PR-4

  • SD-2

  • PD-1

  • Hydroxyurea was safe and elicited durable responses in some patients who had persistent or progressive disease following multiple previous therapies.100

Hematopoietic Cell Transplant 101 Retrospective Total – 87
  • MAC – 20

  • RIC - 67

HSCT with MAC or RIC 3 year OS
  • MAC – 77%

  • RIC – 71%

TRM
  • MAC-28%

  • RIC-8%

  • HSCT offers potential for cure for patients with relapsed and refractory LCH. Myeloablative conditioning is associated with higher treatment-related mortality; reduced intensity conditioning is associated with higher rates of relapse.

For highly refractory high-risk cases, hematopoietic stem cell transplant (HSCT) which replaces the neoplastic clone may be curative. Myeloablative conditioning (MAC) and reduced Intensity (RIC) approaches have similar historic overall survival, though MAC was associated with increased treatment-related mortality and RIC was associated with increased risk of relapse.101 Utilization of HSCT for LCH has decreased significantly as more refractory HR patients are now treated with MAPK inhibition, on- and off-study.

Treatment of Adults with LCH

Adults are frequently intolerant to the pediatric front-line vinblastine/prednisone therapy. Expert opinion therefore recommends methotrexate, cytarabine or cladribine monotherapy for front-line treatment of adults with LCH who require systemic therapy.102,103 Vemurafenib is FDA-approved for treatment of BRAFV600E+ ECD, and cobimetinib is now approved for adults with histiocytic neoplasms. In patients where a quick response is desirable, vemurafenib may be used for BRAFV600E mutated disease. Empiric MEK inhibition is also a reasonable treatment if mutation status is unknown. For adult smokers with isolated pulmonary LCH, smoking cessation may be sufficient. Bisphosphonates or gabapentin may be helpful for LCH-associated pain. A recent study demonstrated an 80% ORR in adults with bone LCH treated with the RANKL inhibitor denosumab.104

LCH-ND

LCH-ND is a highly significant remaining clinical challenge for LCH patients and their physicians due to unpredictable onset and clinical consequences of progressive neurodegeneration. For patients who are cured of systemic LCH, uncertainty of risk for LCH-ND can be a sword of Damocles. As discussed above, recent biological insights demonstrate LCH-ND as a manifestation of clonal LCH.58 Management has historically been aimed at supportive measures including physical therapy and IVIG.38 Clinical series report potential efficacy of LCH-directed therapies including cytarabine105 and vemurafenib;58 however, responses to MAPK inhibition are not as reliable or complete as for systemic LCH. Further, potential for significant clinical improvement may be limited in patients with established brain injury. If LCH-ND arises from hematopoietic precursors, then systematic approaches to prevention (e.g., clinical risk-stratification, peripheral blood BRAFV600E testing, serial MRI imaging) may become important to avoid development of entrenched pathogenic BRAFV600E+ CNS macrophages.31,58

MAPK Pathway Inhibition

Discoveries over the past 13 years have demonstrated that MAPK hyperactivation in myeloid precursors plays a central role in driving LCH lesion formation and pathogenesis. Blocking MAPK hyperactivation therefore offers a rationale “hammer” for this prominent, newly exposed “nail” (Table 5). Adults with LCH and ECD were included in the Memorial Sloan Kettering Phase 2 basket trial for the BRAFV600E inhibitor vemurafenib. Responses were good when measured by RECIST criteria (ORR 61%) and extraordinary when measured by PET-CT (ORR 100%).106,107 Similar experiences were reported in a French series of adults with ECD and mixed ECD/LCH.108 It should be noted that vemurafenib, dabrafenib and other first-generation BRAF inhibitors specifically block BRAFV600E, with paradoxical activation of MAPK signaling in normal cells. This limits the potential patient population and contributes to toxicities.109 However, no such activation accompanies MEK inhibition, where toxicities are related to MAPK attenuation in normal cells. Diamond and colleagues developed a trial to treat “all-comers” with adult histiocytosis with MEK inhibitors based on the rationale that almost all activating mutations associated with histiocytic disorders arise in genes encoding MEK or proximal MAPK pathway genes. This trial demonstrated a metabolic ORR 91% regardless of mutational status in adults and led to FDA approval of cobimetinib for histiocytic disorders in adults. The first child treated with a MAPK inhibitor was reported by Heritier and colleagues in 2015 with dramatic response.110 Subsequent pediatric series supported reproducible high response rates to MAPK inhibitors, even in patients with aggressive disease.83,84,111 Some patients with LCH-ND also responded, though potential for recovery in patients with long-standing disease is perhaps limited by drug penetration of the blood-brain barrier and pre-existing CNS injury.58

Table 5. MAPK inhibition studies for LCH and related conditions.

HR – high risk (risk-organ-positive); LR – low risk (risk-organ negative; LCH-ND – neurodegenerative LCH; ORR – overall response rate; PFS – progression free survival; CR – complete response; PR – partial response.

Study Design Subjects Therapies Response Criteria Response Timepoint Outcomes Conclusions
VE-Basket 106,107 Phase 2 Clinical trial 26 Total - Adults
  • 22 ECD

  • 4 LCH

Vemurafenib RECIST
PERCIST
2–40 months
2–40 months
ORR – 61.5%
ORR – 100%
  • Consistent responses to vemurafenib in adults with BRAF-V600E+ LCH and ECD

  • Frequent but generally manageable toxicities (skin, GI); rare ocular, secondary squamous cell carcinoma.

  • No evidence of acquired resistance to therapy.

  • PET-CT-based responses more sensitive than RECIST.

Targeted Therapy for ECD 108 Retrospective 8 Total - Adults
  • 4 ECD

  • 4 mixed ECD/LCH

Vemurafenib PET response 6 months
10.5 months (median)
ORR-100%
PFS 100%
  • Consistent responses in vemurafenib adults with BRAF-V600E+ LCH and ECD

LOVE Study 112 Retrospective series 54 Total, all ECD Adults
  • 20 in cessation cohort

Various MAPK inhibitors PERCIST 6 months after cessation of MAPK inhibition therapy PFS 25%
  • Rapid relapse in majority of patients with ECD with cessation of MAPK inhibition.

Cobimetinib for Histiocytoses 125 Phase 2 clinical trial 18 total - Adults
  • 12 ECD

  • 2 LCH

  • 2 RDD

  • 2 mixed

Cobimetinib PERCIST 1.6–15.9 months ORR – 88%
  • Cobimetinib (MEK inhibitor) is effective in the majority of adults with histiocytic disorders with a spectrum of driver mutations.

  • This study supported FDA approval for cobimetinib for histiocytic disorders in adults.

Pediatric Case Study 110 Case study 1 HR LCH case - Pediatric Vemurafenib RECIST Day 60 CR
  • 1st pediatric case of severe HR LCH with positive response to vemurafenib.

NACHO-LIBRE 84 Retrospective series 21 LCH - Pediatric
  • 13 LCH-ND without systemic lesions

  • 8 Systemic LCH

MAPK Inhibitors Modified PERCIST 0.6–48 months ORR-86%
  • High response rates in pediatric cohort with LCH-ND and systemic LCH to MAPK inhibition.

  • Persistent peripheral blood mononuclear cell BRAFV600E were detected in patients on MAPK inhibitor therapy, even in patients with positive clinical responses.

European Vemurafenib Study 83 Observational study 54 LCH - Pediatric
  • 44 HR

  • 10 LR

Vemurafenib Disease Activity Score 8 weeks
0.1 – 7.3 months after cessation of vemurafenib
ORR – 100%
  • 38 CR

  • 16 PR

PFS 20%
  • High response rate in pediatric cohort with systemic LCH.

  • Rapid relapse with cessation of therapy

MAPK Inhibitors for Histiocytic Disorders 111 Retrospective series 30 LCH – Pediatric (n=27) and Adult (n=3)
  • 16 front-line

  • 14 salvage

Dabrafenib (10)
Trametinib (17)
Trametinib/Dabrafenib (3)
Histiocyte Society Criteria (including PET-CT) 0.3 month-6.4 years ORR-100%
25/30 (83%) LCH patients achieved NAD without persistent symptoms of disease
  • High response rate to front-line or salvage MAPK inhibition in largely pediatric cohort with LCH.

Dabrafenib and Trametinib Pediatric Study 113 Phase 2 25 LCH – Pediatric
  • 13 Dabrafenib monotherapy

  • 12 Dabrafenib + Trametinib

Dabrafenib
Dabrafenib + Trametinib
Histiocyte Society Criteria 7–65 months
1.8–35.9 months
ORR
  • Dabrafenib – 76.9%

  • Dual therapy – 58.3%

  • Responses likely lower than comparable pediatric cohorts due to HS Response Criteria versus PET-based approaches.

  • Dual therapy did not appear to add benefit in this study.

These early experiences with MAPK inhibitors have been game-changing for some patients with LCH. Death in infants with HR LCH is becoming increasingly rare. However, the LOVE study, in which adults with ECD were followed after achieving complete responses with MAPK inhibitors, signaled a cautionary note with >75% of patients relapsing within 6 months of stopping therapy.112 Similarly the European vemurafenib study also reported rapid relapse in the majority of patients following treatment cessation.83 Unlike other conditions with more complex mutation landscape, resistance to MAPK inhibition does not typically develop in LCH, and patients respond to retreatment with the same MAPK inhibitor following relapse or progression off-therapy. Where combining MAPK inhibitors (e.g. BRAFV600E and MEK inhibitors) have demonstrated superior outcomes in more genomically complex disorders such as melanoma, dabrafenib alone and dabrafenib/trametinib yielded similar outcomes in pediatric LCH patients.113

Chaos and Opportunity

LCH Treatment Approaches: 2024

Prior to 2010, when we had few insights into mechanisms of LCH pathogenesis, there was near global consensus regarding optimal therapeutic approaches for systemic LCH, based on the best results of Histiocyte Society trials. Over the past decade, the pace of discovery has outrun our clinical trial capacity. There are numerous and increasingly available MAPK inhibiting agents that are being used in various settings (e.g. front-line, second-line, beyond), in different types of patients (e.g. LCH, LCH-ND, ECD, RDD, etc.), and in different combinations (single agent, combined targeted agents, combined with cytotoxic chemotherapy, etc.). At the same time, clinical trials to rigorously define responses and toxicities associated with MAPK inhibitor therapy in LCH are limited, especially in children. Therapy recommendations and level of evidence for the range of LCH scenarios are outlined in Table 6. We strongly support enrollment of patients on clinical trials when possible. In the meantime, we favor the conventional approach of using treatment strategies with the best evidence: For front-line therapy, this remains the LCH-III-supported approach of vinblastine/prednisone. However, we acknowledge that in cases where patients are critically ill, such as infants with rapidly progressive multi-system disease and bone marrow and/or liver involvement requiring immediate responses, off-label and off-study use of MAPK inhibition for some period may be reasonable. Standardized treatment recommendations become more difficult for salvage therapy, where data are retrospective and anecdotal.

Table 6.

LCH Treatment Strategies

Front-line Recommendation Evidence
Single bone Curettage +/− local steroid injection (Do NOT aim for gross total resection) Expert opinion
Single bone
  • CNS risk

  • Not surgically accessible/morbidity risk

Vinblastine/prednisone × 12 months (according to LCH-III) Extrapolated from randomized control trial (LCH-III)
Multifocal bone Vinblastine/prednisone × 12 months (according to LCH-III) Extrapolated from randomized control trial (LCH-III)
Multisystem (Low-risk) Vinblastine/prednisone × 12 months (according to LCH-III) Randomized control trial
Multisystem (High-risk) Vinblastine/prednisone/6MP × 12 months (according to LCH-III) Randomized control trial
Multisystem (High-risk)
  • Meets HLH criteria

  • Critically ill

Consider MAPK inhibition to manage acute severe symptoms (ideally on clinical trial) Expert opinion
1st Salvage Recommendation Evidence
Recurrent single bone Vinblastine/prednisone × 12 months – or-
Cytarabine monotherapy (100 mg/m2/day × 5 days/cycle) × 12 months if previous exposure to vinblastine/prednisone.
(If single bone progresses or relapses, we consider this evolution of systemic disease)
Extrapolated from randomized control trial (LCH-III)
Retrospective series
Multifocal bone Cytarabine monotherapy (100 mg/m2/day × 5 days/cycle) × 12
MAPK inhibitor if on clinical trial
Retrospective series
Expert opinion
Multisystem (Low-risk) Cytarabine monotherapy (100 mg/m2/day × 5 days/cycle) × 12
MAPK inhibitor if on clinical trial
Retrospective series
Observational cohort
Multisystem (High-risk) Cytarabine monotherapy (100 mg/m2/day × 5 days/cycle) × 12 -or-
Clofarabine monotherapy (25 mg/m2/day × 5 days/cycle) × 6; if CR at 6 months, then 2 days/cycle × 6 more cycles; if PR at 6 months, then continue 5 day cycles × 6 more cycles. – or -
MAPK inhibitor if on clinical trial; or MAPK inhibitor off-study if disease is rapidly progressive or if patient is critically ill.
Retrospective series
Retrospective series
Observational cohort
2nd Salvage and Beyond Recommendation Evidence
Systemic relapse/progression Clofarabine monotherapy (25 mg/m2/day × 5 days/cycle) × 6; if CR at 6 months, then 2 days/cycle × 6 more cycles; if PR at 6 months, then continue 5 day cycles × 6 more cycles. – or –
MAPK inhibitor, or other therapeutic strategy on clinical trial. – or -
MAPK inhibitor off-study
Expert opinion
Observational cohort
Observational cohort
Special Cases Recommendation Evidence
LCH-ND: LACI Observe, confirm progression of abnormal MRI findings with 2 or more worsening MRI over 3 months
Cytarabine monotherapy (150 mg/m2/day × 5 days/cycle) × 12 -or-
MAPK inhibitor, or other therapeutic strategy on clinical trial. – or -
MAPK inhibitor off-study
Expert opinion
Retrospective series
Observational cohort
Observational cohort
LCH-ND: LACS Cytarabine monotherapy (150 mg/m2/day × 5 days/cycle) × 12 -or-
MAPK inhibitor, or other therapeutic strategy on clinical trial. – or -
MAPK inhibitor off-study
Retrospective series
Observational cohort
Observational cohort
Adult LCH Same as above with exceptions:
Cytarabine or cladribine monotherapy as front-line for systemic disease
MAPK inhibition as 2nd line and greater for systemic disease

Observational cohort
Phase 1–2 clinical trials
Skin-limited LCH Observe
If symptomatic, consider topical steroids and/or oral methotrexate and/or oral hydroxyurea as needed
If persistent/progressive, be sure to image to rule out multi-system LCH
Expert opinion
Systemic disease in lower resource settings Consider oral hydroxyurea or subcutaneous cytarabine Retrospective series

Even with limited data, the efficacy of MAPK inhibitors in most LCH patients seems predictable. However, associated toxicities are unknown. Thus, balancing the costs and benefits for patients with diverse clinical manifestations and across risk groups presents a significant challenge. While acute toxicities are typically manageable, many patients taking MAPK inhibitors experience side effects. Skin rash and GI symptoms are common and severe toxicities including the development of second malignancies have been reported.114 Additionally, identification of other potential side effects of chronic inhibitor therapy, including impact on growth and development in children are not known. Prospective clinical trials with extended follow-up are therefore essential to understand the impact of extended use of MAPK inhibitors in children and adults.115 Traditional chemotherapy also has predictable toxicities, but responses for most patients with LCH do not appear as reliable or rapid compared to MAPK inhibition. However, less than 50% of patients with disseminated LCH are cured with 1 year of vinblastine/prednisone/6MP treatments (48% with “no active disease” status at 1 year in LCHIII).90 Decisions regarding therapy with traditional chemotherapy with the aim for cure versus indefinite management with MAPK inhibition are being made with limited data describing the safety and efficacy of these novel targeted agents. Data on the patient experience with therapy alternatives will be critically important to guide treatment decisions and to optimize quality of life during and after therapy.

The North American Consortium for Histiocytosis (NACHO) consortium developed the NACHO-COBI trial (NCT04079179), to test the safety and efficacy of cobimetinib monotherapy in 1)children and young adults with relapse/refractory LCH; 2)children and adults with LCH-ND; 3)children and young adults with non-LCH histiocytoses (e.g. Rosai-Dorfman disease, juvenile xanthogranuloma, histiocytic sarcoma); and 4)adults with histiocytic disorders (LCH and non-LCH). The first LCH trial in the Children’s Oncology Group (COG) recently opened (ANHL2121, NCT05828069), evaluating the safety and efficacy of a second-generation pan-RAF inhibitor (tovorafenib) in children and young adults with relapsed and refractory LCH. A prospective single center phase 2 trial is testing the MEK inhibitor mirdametinib in newly diagnosed and relapsed and refractory LCH (NCT06153173). While off-label access to MAPK inhibitors in pediatric patients with histiocytic disorders is increasing in the United States, prospective clinical trials are essential to rigorously assess efficacy and toxicity and establish standards of care. Importantly, these studies will accelerate our understanding of biological and clinical correlates of disease response, resistance and outcomes.

Opportunities

After decades of incremental advances, mechanistic insights now offer footholds for opportunities to optimize therapies (and cures) for every patient with LCH.

Risk stratification.

Histiocyte Society clinical trials identified patients at highest risk of death from LCH based on presenting sites of disease.37,90 Molecular data may now inform refined risk stratification strategies. For example, somatic BRAFV600E mutations in LCH lesions have been associated with increased risks of systemic disease and LCH-ND.14,58,59 Further, detection of BRAFV600E in blood via cell-free DNA or in PBMCs is also associated with systemic disease, with BRAFV600E allele frequency tracking disease burden in patients treated with chemotherapy.14,82 Custom ddPCR primers can be used to characterize similar patterns in patients with alternative MAPK pathway mutations.116 The limited correlation between clinical responses and BRAFV600E allele frequency in blood in patients treated with MAPK inhibition is consistent with rapid relapse with therapy cessation.83,84 MAPK inhibitors therefore appear to be set to “stun” rather than “kill”, suppressing the distal pathway of LCH development rather than eliminating the precursor pool. Inclusion of “minimal detectable disease” and “minimal residual disease” strategies on prospective clinical trials will help develop valuable risk stratification tools to guide therapy. Further, the ability to predict risk of LCH-ND may guide surveillance strategies.

New Agents and Combinations.

After decades of vinblastine and prednisone in pediatric disease, our current challenge is prioritizing the clinical testing of dozens of novel agents with potential efficacy in LCH. For most drugs currently used in LCH salvage, the best available data are limited to case series. The general signal of MAPK inhibitor activity in LCH is revolutionary, but we do not know how distinct pharmacodynamic features or distribution (e.g., CNS penetration) impact efficacy, toxicity and durability of response. Beyond chemotherapy and MAPK inhibitors, emerging biology supports the potential for alternative therapeutic strategies including targeting the survival advantage induced by MAPK activation (e.g., apoptosis dis-inhibition with BH3 mimetics)30, other senolytic agents to inhibit the senescence-associated secretory phenotype (e.g. sirolimus)30, differentiation blockade (e.g., HDAC inhibitors)14, antigen targets (e.g. CSF1R, CD207, SIRPα)117,118, and immune checkpoint inhibitors (e.g. PD-1)119. Chemotherapy for LCH is not always effective but provides the chance of cure, whereas MAPK inhibition is highly active but does not seem to cure. Therefore, identifying how to most effectively combine agents to achieve safe, reliable and durable cure is a key priority. Combining intermediate-dose cytarabine/cladribine with vemurafenib was feasible in a prospective cohort (n=19) with 2-year “reactivation/progression-free survival” (RFS) 76.9% for risk-organ+ (RO+) and 83.3% for RO-; one patient developed myelodysplastic syndrome with uncertain relationship to therapy120. Pre-clinical models (primary cell culture and mice) may be able to contribute to drug and drug combination prioritization.30,33

Quality of Life/ Survivorship.

There are now many paths to prevent death in patients with LCH. Current front-line practices range from a year of chemotherapy to a life of MAPK inhibition. Which is better? Current response evaluations are focused on ORR and PFS. If potential for death has been largely eliminated, clinical trials may not answer the most important questions for patients regarding their experiences on therapy and after. Building patient-reported quality of life data into clinical trials will be essential to understand which approaches are the “best”. Similarly, systematic collection of epidemiologic data and long-term outcomes is required to understand the life-long impacts of this disease and different treatments.

Access to Therapy.

Most patients in the world who develop histiocytic disorders do not have access to centers or drugs required for chemotherapy infusions or targeted oral therapies. Oral hydroxyurea offers an opportunity to treat patients with “smoldering” disease – and may offer insights into the relative benefit of duration of cytotoxic therapy against myeloid precursors versus intensity.100 As another option for rural or lower-resource settings, cytarabine may be given subcutaneously, avoiding need for frequent IV access or port placement.121

Organizational Collaboration and Standardized Endpoints.

Prospective clinical trials are essential to identify optimal therapies for LCH. NACHO was created to catalyze improvements in outcomes for patients with histiocytic disorders. We grew from 11 original members in 2014 to 65 current member institutions. This growth was not initially planned or expected, reflecting the great interest in participating in LCH research and previously unmet need. Initiation of the European Consortium for Histiocytosis (ECHO) and other regional/national groups, and inclusion of LCH in COG are tremendous opportunities to advance research. Collaboration and coordination are essential to amplify the impact of research organizations where multiple complementary studies can be conducted in parallel. Harmonized endpoints and common data dictionaries will be important to facilitate comparisons between studies.

Conclusions

After more than 100 years of developing therapeutic strategies for patients with LCH in the “dark”, we now have the challenge of testing, prioritizing and implementing strategies based on mechanistic insights that are becoming rapidly illuminated. In comparison to the frenetic pace of biological discoveries, the clinical trial and regulatory environments have limited acceleration capacity. We therefore have the challenge to strategically prioritize and test novel concepts as efficiently and effectively as possible. In the meantime, as patients continue to arrive in our offices, we rely on conventional approaches to evaluate existing clinical data and contribute patient experiences to cohorts and trials whenever possible. Given that many new therapies now effectively avoid death and produce unprecedented response rates, the voices of our patients and families are increasingly important to inform the best experiences during and after therapy. Our goal over the next decade is to identify and implement the best possible therapy for each patient with LCH to ensure long-term survival with optimal quality of life.

Table 2.

Differential diagnoses and LCH.

Area of Involvement Clinical Manifestation Possible Differential Diagnoses
Skin Dermatitis Seborrheic dermatitis
Vesicles Varicella, herpes simplex, erythema toxicum
Petechiae Immune thrombocytopenia, leukemia
Ulcerative lesions Fungal infection
Nodules Juvenile xanthogranuloma, infant leukemia, mastocytosis, neuroblastoma, subcutaneous panniculitis-like T cell lymphoma
Bone Lytic lesions/vertebra plana Acute osteomyelitis, Chronic relapsing multifocal osteomyelitis, Atypical mycobacterial infection, Bone angiomatosis (Gorham disease), Aneurysmal bone cyst, Juvenile xanthogranuloma, Malignancy
Lung Cavitary nodules Mycobacterial or other infection, sarcoidosis
Pneumocystis jirovecii
Liver Jaundice/hypoalbuminemia Hepatitis, metabolic disease, malignancy, chronic active EBV, NK/T cell lymphoma, toxic injury
Pituitary Diabetes insipidus Central nervous system germ cell tumor, hypophysitis

Acknowledgements:

NACHO (CEA, MM, CRG) receives grant support from the Leukemia and Lymphoma Society Translational Research Program, from the St. Baldrick’s Foundation Consortium Grant through the Moves for Miles Childhood Cancer Foundation. This work was supported by the National Institutes of Health/National Cancer Institute (P30 CA008748; ELD, ID) as well as the National Cancer Institute (R37CA259260; ELD). This was supported by the Frame Family Fund (ELD), the Joy Family West Foundation (ELD), and the Applebaum Foundation (ELD). CEA and MM receive funding from the National Institute of Health (R01 CA154947). CEA, OSE, NEM, JL and KLM are supported by the HistioCure Foundation and Judy and Henry Sauer. We appreciate colleagues at HistioUK who collaborated on development and review of this manuscript.

Conflicts:

AK has participated in consulting and/or advisory boards for Sobi and SpringWorks Therapeutics.

CEA has participated in consulting and/or advisory boards for Sobi, OPNA, Electra, and receives research support from Genentech.

MMH has participated in consulting and/or advisory boards for Sobi and Electra.

GG has participated on advisory boards for OPNA and SeaGen.

ELD discloses unpaid editorial support from Pfizer Inc and serves on an advisory board for Day One Therapeutics, Opna Bio, and Springworks Therapeutics, both outside the submitted work.

References

  • 1.Langerhans P Ueber die Nerven der menschlichen Haut. Virchows Archives der pathologischen Anatomie. 1868;44:325. [Google Scholar]
  • 2.Langerhans P. Berichtigungen. Archiv der mikroskopischen Anatomie. 1882;20:641. [Google Scholar]
  • 3.Hand AJ. Polyuria and Tuberculosis. Archives of Pediatrics. 1893;10:673. [Google Scholar]
  • 4.Schueller A Ueber eigenartige Schaedeldefekte im Jugendalter. Fortschritte der Roentgenstrahlen. 1915;23:12. [Google Scholar]
  • 5.Christian HA. Defects in membraneous bones, exophathalmos and diabetes insipidus: an unusual syndrome of dyspituitarism. Medical Clinics of North America. 1919;3:849. [Google Scholar]
  • 6.Letterer E Aleukaemische Retikulocytose (Ein Beitrag zu den proliferativen Erkrankungen des Retikuloendothelialapparates). Frankfurter Zeitschrift fuer die Pathologie. 1924;30(377) [Google Scholar]
  • 7.Siwe SA. Die Reikuloendotheliose-ein neues Krankheitsblid unter den Hepatosplenomegalien. Zur Kinderheilkunde. 1933;55:212. [Google Scholar]
  • 8.Farber S The nature of “solitary of eosinophilic granuloma” of bone. American Journal of Pathology. 1944;17:625. [Google Scholar]
  • 9.Lichtenstein L Histiocytosis X; integration of eosinophilic granuloma of bone, Letterer-Siwe disease, and Schuller-Christian disease as related manifestations of a single nosologic entity. AMA Arch Pathol. Jul 1953;56(1):84–102. [PubMed] [Google Scholar]
  • 10.Nezelof C, Basset F, Rousseau MF. Histiocytosis X histogenetic arguments for a Langerhans cell origin. Biomedicine. Sep 1973;18(5):365–71. [PubMed] [Google Scholar]
  • 11.Willman CL, Busque L, Griffith BB, et al. Langerhans’-cell histiocytosis (histiocytosis X)--a clonal proliferative disease. N Engl J Med. Jul 21 1994;331(3):154–60. doi: 10.1056/NEJM199407213310303 [DOI] [PubMed] [Google Scholar]
  • 12.Yu RC, Chu C, Buluwela L, Chu AC. Clonal proliferation of Langerhans cells in Langerhans cell histiocytosis. Lancet. Mar 26 1994;343(8900):767–8. [DOI] [PubMed] [Google Scholar]
  • 13.Badalian-Very G, Vergilio JA, Degar BA, et al. Recurrent BRAF mutations in Langerhans cell histiocytosis. Blood. Sep 16 2010;116(11):1919–23. doi: 10.1182/blood-2010-04-279083 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Berres ML, Lim KP, Peters T, et al. BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups. J Exp Med. Apr 7 2014;211(4):669–83. doi: 10.1084/jem.20130977 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Berres ML, Merad M, Allen CE. Progress in understanding the pathogenesis of Langerhans cell histiocytosis: back to Histiocytosis X? Br J Haematol. Apr 2015;169(1):3–13. doi: 10.1111/bjh.13247 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Arceci RJ, Brenner MK, Pritchard J. Controversies and new approaches to treatment of Langerhans cell histiocytosis. Hematol Oncol Clin North Am. Apr 1998;12(2):339–57. doi: 10.1016/s0889-8588(05)70514-1 [DOI] [PubMed] [Google Scholar]
  • 17.Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia. Jul 2022;36(7):1720–1748. doi: 10.1038/s41375-022-01620-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Picarsic J, Jaffe R. Nosology and Pathology of Langerhans Cell Histiocytosis. Hematol Oncol Clin North Am. Oct 2015;29(5):799–823. doi: 10.1016/j.hoc.2015.06.001 [DOI] [PubMed] [Google Scholar]
  • 19.Chikwava K, Jaffe R. Langerin (CD207) staining in normal pediatric tissues, reactive lymph nodes, and childhood histiocytic disorders. Pediatr Dev Pathol. Nov–Dec 2004;7(6):607–14. doi: 10.1007/s10024-004-3027-z [DOI] [PubMed] [Google Scholar]
  • 20.Favara BE, Feller AC, Pauli M, et al. Contemporary classification of histiocytic disorders. The WHO Committee On Histiocytic/Reticulum Cell Proliferations. Reclassification Working Group of the Histiocyte Society. Med Pediatr Oncol. Sep 1997;29(3):157–66. doi: [DOI] [PubMed] [Google Scholar]
  • 21.Histiocytosis syndromes in children. Writing Group of the Histiocyte Society. Lancet. Jan 24 1987;1(8526):208–9. [PubMed] [Google Scholar]
  • 22.Guilliams M, Ginhoux F, Jakubzick C, et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol. Aug 2014;14(8):571–8. doi: 10.1038/nri3712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Emile JF, Abla O, Fraitag S, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. Jun 2 2016;127(22):2672–81. doi: 10.1182/blood-2016-01-690636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Khoury JD, Solary E, Abla O, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia. Jul 2022;36(7):1703–1719. doi: 10.1038/s41375-022-01613-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chakraborty R, Hampton OA, Shen X, et al. Mutually exclusive recurrent somatic mutations in MAP2K1 and BRAF support a central role for ERK activation in LCH pathogenesis. Blood. Nov 6 2014;124(19):3007–15. doi: 10.1182/blood-2014-05-577825 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chakraborty R, Hampton OA, Abhyankar H, et al. Activating MAPK1 (ERK2) mutation in an aggressive case of disseminated juvenile xanthogranuloma. Oncotarget. Jul 11 2017;8(28):46065–46070. doi: 10.18632/oncotarget.17521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Diamond EL, Durham BH, Haroche J, et al. Diverse and Targetable Kinase Alterations Drive Histiocytic Neoplasms. Cancer Discov. Feb 2016;6(2):154–65. doi: 10.1158/2159-8290.CD-15-0913 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lavoie H, Gagnon J, Therrien M. ERK signalling: a master regulator of cell behaviour, life and fate. Nat Rev Mol Cell Biol. Oct 2020;21(10):607–632. doi: 10.1038/s41580-020-0255-7 [DOI] [PubMed] [Google Scholar]
  • 29.Poulikakos PI, Sullivan RJ, Yaeger R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin Cancer Res. Nov 1 2022;28(21):4618–4628. doi: 10.1158/1078-0432.CCR-21-2138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bigenwald C, Le Berichel J, Wilk CM, et al. BRAF(V600E)-induced senescence drives Langerhans cell histiocytosis pathophysiology. Nat Med. May 2021;27(5):851–861. doi: 10.1038/s41591-021-01304-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wilk CM, Cathomas F, Torok O, et al. Circulating senescent myeloid cells infiltrate the brain and cause neurodegeneration in histiocytic disorders. Immunity. Dec 12 2023;56(12):2790–2802 e6. doi: 10.1016/j.immuni.2023.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Allen CE, Merad M, McClain KL. Langerhans-Cell Histiocytosis. N Engl J Med. Aug 30 2018;379(9):856–868. doi: 10.1056/NEJMra1607548 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Hogstad B, Berres ML, Chakraborty R, et al. RAF/MEK/extracellular signal-related kinase pathway suppresses dendritic cell migration and traps dendritic cells in Langerhans cell histiocytosis lesions. J Exp Med. Jan 2 2018;215(1):319–336. doi: 10.1084/jem.20161881 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Peckham-Gregory EC, Chakraborty R, Scheurer ME, et al. A genome-wide association study of LCH identifies a variant in SMAD6 associated with susceptibility. Blood. Nov 16 2017;130(20):2229–2232. doi: 10.1182/blood-2017-08-800565 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Ribeiro KB, Degar B, Antoneli CB, Rollins B, Rodriguez-Galindo C. Ethnicity, race, and socioeconomic status influence incidence of Langerhans cell histiocytosis. Pediatr Blood Cancer. Jun 2015;62(6):982–7. doi: 10.1002/pbc.25404 [DOI] [PubMed] [Google Scholar]
  • 36.Goyal G, Shah MV, Hook CC, et al. Adult disseminated Langerhans cell histiocytosis: incidence, racial disparities and long-term outcomes. Br J Haematol. Aug 2018;182(4):579–581. doi: 10.1111/bjh.14818 [DOI] [PubMed] [Google Scholar]
  • 37.Gadner H, Grois N, Potschger U, et al. Improved outcome in multisystem Langerhans cell histiocytosis is associated with therapy intensification. Blood. Mar 1 2008;111(5):2556–62. doi: 10.1182/blood-2007-08-106211 [DOI] [PubMed] [Google Scholar]
  • 38.Yeh EA, Greenberg J, Abla O, et al. Evaluation and treatment of Langerhans cell histiocytosis patients with central nervous system abnormalities: Current views and new vistas. Pediatr Blood Cancer. Jan 2018;65(1)doi: 10.1002/pbc.26784 [DOI] [PubMed] [Google Scholar]
  • 39.Goyal G, Acosta-Medina AA, Abeykoon JP, et al. Long-term outcomes among adults with Langerhans cell histiocytosis. Blood Adv. Nov 14 2023;7(21):6568–6578. doi: 10.1182/bloodadvances.2023010706 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Simko SJ, Garmezy B, Abhyankar H, et al. Differentiating skin-limited and multisystem Langerhans cell histiocytosis. J Pediatr. Nov 2014;165(5):990–6. doi: 10.1016/j.jpeds.2014.07.063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Munn S, Chu AC. Langerhans cell histiocytosis of the skin. Hematol Oncol Clin North Am. Apr 1998;12(2):269–86. [DOI] [PubMed] [Google Scholar]
  • 42.Ashena Z, Alavi S, Arzanian MT, Eshghi P. Nail involvement in langerhans cell histiocytosis. Pediatr Hematol Oncol. Jan–Feb 2007;24(1):45–51. doi: 10.1080/08880010601001362 [DOI] [PubMed] [Google Scholar]
  • 43.Madrigal-Martinez-Pereda C, Guerrero-Rodriguez V, Guisado-Moya B, Meniz-Garcia C. Langerhans cell histiocytosis: literature review and descriptive analysis of oral manifestations. Med Oral Patol Oral Cir Bucal. May 1 2009;14(5):E222–8. [PubMed] [Google Scholar]
  • 44.Slater JM, Swarm OJ. Eosinophilic granuloma of bone. Med Pediatr Oncol. 1980;8(2):151–64. [DOI] [PubMed] [Google Scholar]
  • 45.Peng XS, Pan T, Chen LY, Huang G, Wang J. Langerhans’ cell histiocytosis of the spine in children with soft tissue extension and chemotherapy. Int Orthop. Jun 2009;33(3):731–6. doi: 10.1007/s00264-008-0529-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ronceray L, Potschger U, Janka G, et al. Pulmonary involvement in pediatric-onset multisystem Langerhans cell histiocytosis: effect on course and outcome. J Pediatr. Jul 2012;161(1):129–33 e1–3. doi: 10.1016/j.jpeds.2011.12.035 [DOI] [PubMed] [Google Scholar]
  • 47.Abbritti M, Mazzei MA, Bargagli E, et al. Utility of spiral CAT scan in the follow-up of patients with pulmonary Langerhans cell histiocytosis. Eur J Radiol. Aug 2012;81(8):1907–12. doi: 10.1016/j.ejrad.2011.04.018 [DOI] [PubMed] [Google Scholar]
  • 48.Bernstrand C, Cederlund K, Henter JI. Pulmonary function testing and pulmonary Langerhans cell histiocytosis. Pediatr Blood Cancer. Sep 2007;49(3):323–8. doi: 10.1002/pbc.20707 [DOI] [PubMed] [Google Scholar]
  • 49.Ducassou S, Seyrig F, Thomas C, et al. Thymus and mediastinal node involvement in childhood Langerhans cell histiocytosis: long-term follow-up from the French national cohort. Pediatr Blood Cancer. Nov 2013;60(11):1759–65. doi: 10.1002/pbc.24603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Grois NG, Favara BE, Mostbeck GH, Prayer D. Central nervous system disease in Langerhans cell histiocytosis. Hematol Oncol Clin North Am. Apr 1998;12(2):287–305. [DOI] [PubMed] [Google Scholar]
  • 51.Marchand I, Barkaoui MA, Garel C, Polak M, Donadieu J, Writing C. Central diabetes insipidus as the inaugural manifestation of Langerhans cell histiocytosis: natural history and medical evaluation of 26 children and adolescents. J Clin Endocrinol Metab. Sep 2011;96(9):E1352–60. doi: 10.1210/jc.2011-0513 [DOI] [PubMed] [Google Scholar]
  • 52.Dunger DB, Broadbent V, Yeoman E, et al. The frequency and natural history of diabetes insipidus in children with Langerhans-cell histiocytosis. N Engl J Med. Oct 26 1989;321(17):1157–62. doi: 10.1056/NEJM198910263211704 [DOI] [PubMed] [Google Scholar]
  • 53.Prosch H, Grois N, Prayer D, et al. Central diabetes insipidus as presenting symptom of Langerhans cell histiocytosis. Pediatr Blood Cancer. Oct 2004;43(5):594–9. doi: 10.1002/pbc.20102 [DOI] [PubMed] [Google Scholar]
  • 54.Grois N, Potschger U, Prosch H, et al. Risk factors for diabetes insipidus in langerhans cell histiocytosis. Pediatr Blood Cancer. Feb 2006;46(2):228–33. doi: 10.1002/pbc.20425 [DOI] [PubMed] [Google Scholar]
  • 55.Prayer D, Grois N, Prosch H, Gadner H, Barkovich AJ. MR imaging presentation of intracranial disease associated with Langerhans cell histiocytosis. AJNR Am J Neuroradiol. May 2004;25(5):880–91. [PMC free article] [PubMed] [Google Scholar]
  • 56.Donadieu J, Rolon MA, Thomas C, et al. Endocrine involvement in pediatric-onset Langerhans’ cell histiocytosis: a population-based study. J Pediatr. Mar 2004;144(3):344–50. doi: 10.1016/j.jpeds.2003.12.030 [DOI] [PubMed] [Google Scholar]
  • 57.Fahrner B, Prosch H, Minkov M, et al. Long-term outcome of hypothalamic pituitary tumors in Langerhans cell histiocytosis. Pediatr Blood Cancer. Apr 2012;58(4):606–10. doi: 10.1002/pbc.24042 [DOI] [PubMed] [Google Scholar]
  • 58.McClain KL, Picarsic J, Chakraborty R, et al. CNS Langerhans cell histiocytosis: Common hematopoietic origin for LCH-associated neurodegeneration and mass lesions. Cancer. Jun 15 2018;124(12):2607–2620. doi: 10.1002/cncr.31348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Heritier S, Barkaoui MA, Miron J, et al. Incidence and risk factors for clinical neurodegenerative Langerhans cell histiocytosis: a longitudinal cohort study. Br J Haematol. Nov 2018;183(4):608–617. doi: 10.1111/bjh.15577 [DOI] [PubMed] [Google Scholar]
  • 60.Wnorowski M, Prosch H, Prayer D, Janssen G, Gadner H, Grois N. Pattern and course of neurodegeneration in Langerhans cell histiocytosis. J Pediatr. Jul 2008;153(1):127–32. doi: 10.1016/j.jpeds.2007.12.042 [DOI] [PubMed] [Google Scholar]
  • 61.Mittheisz E, Seidl R, Prayer D, et al. Central nervous system-related permanent consequences in patients with Langerhans cell histiocytosis. Pediatr Blood Cancer. Jan 2007;48(1):50–6. doi: 10.1002/pbc.20760 [DOI] [PubMed] [Google Scholar]
  • 62.Milne P, Bomken S, Slater O, et al. Lineage switching of the cellular distribution of BRAFV600E in multisystem Langerhans cell histiocytosis. Blood Adv. May 23 2023;7(10):2171–2176. doi: 10.1182/bloodadvances.2021006732 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.McClain K, Ramsay NK, Robison L, Sundberg RD, Nesbit M, Jr. Bone marrow involvement in histiocytosis X. Med Pediatr Oncol. 1983;11(3):167–71. [DOI] [PubMed] [Google Scholar]
  • 64.Minkov M, Potschger U, Grois N, Gadner H, Dworzak MN. Bone marrow assessment in Langerhans cell histiocytosis. Pediatr Blood Cancer. Oct 15 2007;49(5):694–8. doi: 10.1002/pbc.21227 [DOI] [PubMed] [Google Scholar]
  • 65.Galluzzo ML, Braier J, Rosenzweig SD, Garcia de Davila MT, Rosso D. Bone marrow findings at diagnosis in patients with multisystem langerhans cell histiocytosis. Pediatr Dev Pathol. Mar–Apr 2010;13(2):101–6. doi: 10.2350/09-05-0651-OA.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ballester LY, Cantu MD, Lim KPH, et al. The use of BRAF V600E mutation-specific immunohistochemistry in pediatric Langerhans cell histiocytosis. Hematol Oncol. Feb 2018;36(1):307–315. doi: 10.1002/hon.2388 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Wong A, Ortiz-Neira CL, Reslan WA, et al. Liver involvement in Langerhans cell histiocytosis. Pediatr Radiol. Oct 2006;36(10):1105–7. doi: 10.1007/s00247-006-0262-7 [DOI] [PubMed] [Google Scholar]
  • 68.Jaffe R Liver involvement in the histiocytic disorders of childhood. Pediatr Dev Pathol. May–Jun 2004;7(3):214–25. doi: 10.1007/s10024-003-9876-z [DOI] [PubMed] [Google Scholar]
  • 69.Braier J, Ciocca M, Latella A, de Davila MG, Drajer M, Imventarza O. Cholestasis, sclerosing cholangitis, and liver transplantation in Langerhans cell Histiocytosis. Med Pediatr Oncol. Mar 2002;38(3):178–82. doi: 10.1002/mpo.1306 [DOI] [PubMed] [Google Scholar]
  • 70.Hadzic N, Pritchard J, Webb D, et al. Recurrence of Langerhans cell histiocytosis in the graft after pediatric liver transplantation. Transplantation. Sep 15 2000;70(5):815–9. [DOI] [PubMed] [Google Scholar]
  • 71.Geissmann F, Thomas C, Emile JF, et al. Digestive tract involvement in Langerhans cell histiocytosis. The French Langerhans Cell Histiocytosis Study Group. J Pediatr. Dec 1996;129(6):836–45. [DOI] [PubMed] [Google Scholar]
  • 72.Hait E, Liang M, Degar B, Glickman J, Fox VL. Gastrointestinal tract involvement in Langerhans cell histiocytosis: case report and literature review. Pediatrics. Nov 2006;118(5):e1593–9. doi: 10.1542/peds.2006-0708 [DOI] [PubMed] [Google Scholar]
  • 73.Minkov M, Potschger U, Thacker N, et al. Additive Prognostic Impact of Gastrointestinal Involvement in Severe Multisystem Langerhans Cell Histiocytosis. J Pediatr. Oct 2021;237:65–70 e3. doi: 10.1016/j.jpeds.2021.06.016 [DOI] [PubMed] [Google Scholar]
  • 74.Burnett A, Carney D, Mukhopadhyay S, Scalzetti EM, Leino D, Souid AK. Thyroid involvement with Langerhans cell histiocytosis in a 3-year-old male. Pediatr Blood Cancer. Mar 2008;50(3):726–7. doi: 10.1002/pbc.21030 [DOI] [PubMed] [Google Scholar]
  • 75.Boztug K, Frimpong-Ansah K, Nanduri VR, Lawson J, Russell-Eggitt I, Brock P. Intraocular Langerhans cell histiocytosis in a neonate resulting in bilateral loss of vision. Pediatr Blood Cancer. Oct 15 2006;47(5):633–5. doi: 10.1002/pbc.20714 [DOI] [PubMed] [Google Scholar]
  • 76.Goyal G, Heaney ML, Collin M, et al. Erdheim-Chester disease: consensus recommendations for evaluation, diagnosis, and treatment in the molecular era. Blood. May 28 2020;135(22):1929–1945. doi: 10.1182/blood.2019003507 [DOI] [PubMed] [Google Scholar]
  • 77.Papo M, Diamond EL, Cohen-Aubart F, et al. High prevalence of myeloid neoplasms in adults with non-Langerhans cell histiocytosis. Blood. Aug 24 2017;130(8):1007–1013. doi: 10.1182/blood-2017-01-761718 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hervier B, Haroche J, Arnaud L, et al. Association of both Langerhans cell histiocytosis and Erdheim-Chester disease linked to the BRAFV600E mutation. Blood. Aug 14 2014;124(7):1119–26. doi: 10.1182/blood-2013-12-543793 [DOI] [PubMed] [Google Scholar]
  • 79.Reiner AS, Bossert D, Buthorn JJ, et al. Patient-reported fatigue and pain in Erdheim-Chester disease: a registry-based, mixed methods study. Haematologica. Jun 1 2023;108(6):1685–1690. doi: 10.3324/haematol.2022.282287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Phillips M, Allen C, Gerson P, McClain K. Comparison of FDG-PET scans to conventional radiography and bone scans in management of Langerhans cell histiocytosis. Pediatr Blood Cancer. Jan 2009;52(1):97–101. doi: 10.1002/pbc.21782 [DOI] [PubMed] [Google Scholar]
  • 81.Baratto L, Nyalakonda R, Theruvath AJ, et al. Comparison of whole-body DW-MRI with 2-[(18)F]FDG PET for staging and treatment monitoring of children with Langerhans cell histiocytosis. Eur J Nucl Med Mol Imaging. May 2023;50(6):1689–1698. doi: 10.1007/s00259-023-06122-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Hyman DM, Diamond EL, Vibat CR, et al. Prospective blinded study of BRAFV600E mutation detection in cell-free DNA of patients with systemic histiocytic disorders. Cancer Discov. Jan 2015;5(1):64–71. doi: 10.1158/2159-8290.CD-14-0742 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Donadieu J, Larabi IA, Tardieu M, et al. Vemurafenib for Refractory Multisystem Langerhans Cell Histiocytosis in Children: An International Observational Study. J Clin Oncol. Nov 1 2019;37(31):2857–2865. doi: 10.1200/JCO.19.00456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Eckstein OS, Visser J, Rodriguez-Galindo C, Allen CE, Group N-LS. Clinical responses and persistent BRAF V600E(+) blood cells in children with LCH treated with MAPK pathway inhibition. Blood. Apr 11 2019;133(15):1691–1694. doi: 10.1182/blood-2018-10-878363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Allen CE, Ladisch S, McClain KL. How I treat Langerhans cell histiocytosis. Blood. Jul 2 2015;126(1):26–35. doi: 10.1182/blood-2014-12-569301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Cohen M, Zornoza J, Cangir A, Murray JA, Wallace S. Direct injection of methylprednisolone sodium succinate in the treatment of solitary eosinophilic granuloma of bone: a report of 9 cases. Radiology. Aug 1980;136(2):289–93. doi: 10.1148/radiology.136.2.7403498 [DOI] [PubMed] [Google Scholar]
  • 87.Steinbok P, Cheong A, Dix DB, et al. Nonoperative Management of Childhood Calvarial Langerhans-Cell Histiocytosis. N Engl J Med. Jun 30 2022;386(26):2532–2534. doi: 10.1056/NEJMc2203820 [DOI] [PubMed] [Google Scholar]
  • 88.Minkov M, Grois N, Heitger A, Potschger U, Westermeier T, Gadner H. Treatment of multisystem Langerhans cell histiocytosis. Results of the DAL-HX 83 and DAL-HX 90 studies. DAL-HX Study Group. Klin Padiatr. Jul–Aug 2000;212(4):139–44. doi: 10.1055/s-2000-9667 [DOI] [PubMed] [Google Scholar]
  • 89.Gadner H, Grois N, Arico M, et al. A randomized trial of treatment for multisystem Langerhans’ cell histiocytosis. J Pediatr. May 2001;138(5):728–34. doi: 10.1067/mpd.2001.111331 [DOI] [PubMed] [Google Scholar]
  • 90.Gadner H, Minkov M, Grois N, et al. Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis. Blood. Jun 20 2013;121(25):5006–14. doi: 10.1182/blood-2012-09-455774 [DOI] [PubMed] [Google Scholar]
  • 91.Morimoto A, Ikushima S, Kinugawa N, et al. Improved outcome in the treatment of pediatric multifocal Langerhans cell histiocytosis: Results from the Japan Langerhans Cell Histiocytosis Study Group-96 protocol study. Cancer. Aug 1 2006;107(3):613–9. doi: 10.1002/cncr.21985 [DOI] [PubMed] [Google Scholar]
  • 92.Egeler RM, de Kraker J, Voute PA. Cytosine-arabinoside, vincristine, and prednisolone in the treatment of children with disseminated Langerhans cell histiocytosis with organ dysfunction: experience at a single institution. Med Pediatr Oncol. 1993;21(4):265–70. doi: 10.1002/mpo.2950210406 [DOI] [PubMed] [Google Scholar]
  • 93.Braier J, Rosso D, Pollono D, et al. Symptomatic bone langerhans cell histiocytosis treated at diagnosis or after reactivation with indomethacin alone. J Pediatr Hematol Oncol. Jul 2014;36(5):e280–4. doi: 10.1097/MPH.0000000000000165001 [DOI] [PubMed] [Google Scholar]
  • 94.Weitzman S, Braier J, Donadieu J, et al. 2’-Chlorodeoxyadenosine (2-CdA) as salvage therapy for Langerhans cell histiocytosis (LCH). results of the LCH-S-98 protocol of the Histiocyte Society. Pediatr Blood Cancer. Dec 15 2009;53(7):1271–6. doi: 10.1002/pbc.22229 [DOI] [PubMed] [Google Scholar]
  • 95.Donadieu J, Bernard F, van Noesel M, et al. Cladribine and cytarabine in refractory multisystem Langerhans cell histiocytosis: results of an international phase 2 study. Blood. Sep 17 2015;126(12):1415–23. doi: 10.1182/blood-2015-03-635151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Simko SJ, McClain KL, Allen CE. Up-front therapy for LCH: is it time to test an alternative to vinblastine/prednisone? Br J Haematol. Apr 2015;169(2):299–301. doi: 10.1111/bjh.13208 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Abraham A, Alsultan A, Jeng M, Rodriguez-Galindo C, Campbell PK. Clofarabine salvage therapy for refractory high-risk langerhans cell histiocytosis. Pediatr Blood Cancer. Jun 2013;60(6):E19–22. doi: 10.1002/pbc.24436 [DOI] [PubMed] [Google Scholar]
  • 98.Rodriguez-Galindo C, Jeng M, Khuu P, McCarville MB, Jeha S. Clofarabine in refractory Langerhans cell histiocytosis. Pediatr Blood Cancer. Nov 2008;51(5):703–6. doi: 10.1002/pbc.21668 [DOI] [PubMed] [Google Scholar]
  • 99.Simko SJ, Tran HD, Jones J, et al. Clofarabine salvage therapy in refractory multifocal histiocytic disorders, including Langerhans cell histiocytosis, juvenile xanthogranuloma and Rosai-Dorfman disease. Pediatr Blood Cancer. Mar 2014;61(3):479–87. doi: 10.1002/pbc.24772 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Zinn DJ, Grimes AB, Lin H, Eckstein O, Allen CE, McClain KL. Hydroxyurea: a new old therapy for Langerhans cell histiocytosis. Blood. Nov 17 2016;128(20):2462–2465. doi: 10.1182/blood-2016-06-721993 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Veys PA, Nanduri V, Baker KS, et al. Haematopoietic stem cell transplantation for refractory Langerhans cell histiocytosis: outcome by intensity of conditioning. Br J Haematol. Jun 2015;169(5):711–8. doi: 10.1111/bjh.13347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Goyal G, Tazi A, Go RS, et al. International expert consensus recommendations for the diagnosis and treatment of Langerhans cell histiocytosis in adults. Blood. Apr 28 2022;139(17):2601–2621. doi: 10.1182/blood.2021014343 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Go RS, Jacobsen E, Baiocchi R, et al. Histiocytic Neoplasms, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. Nov 2021;19(11):1277–1303. doi: 10.6004/jnccn.2021.0053 [DOI] [PubMed] [Google Scholar]
  • 104.Makras P, Yavropoulou MP, Chatziioannou SN, et al. Efficacy of denosumab monotherapy among adults with Langerhans cell histiocytosis: A prospective clinical trial. Am J Hematol. Jul 2023;98(7):E168–E171. doi: 10.1002/ajh.26936 [DOI] [PubMed] [Google Scholar]
  • 105.Allen CE, Flores R, Rauch R, et al. Neurodegenerative central nervous system Langerhans cell histiocytosis and coincident hydrocephalus treated with vincristine/cytosine arabinoside. Pediatr Blood Cancer. Mar 2010;54(3):416–23. doi: 10.1002/pbc.22326 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Diamond EL, Subbiah V, Lockhart AC, et al. Vemurafenib for BRAF V600-Mutant Erdheim-Chester Disease and Langerhans Cell Histiocytosis: Analysis of Data From the Histology-Independent, Phase 2, Open-label VE-BASKET Study. JAMA Oncol. Mar 1 2018;4(3):384–388. doi: 10.1001/jamaoncol.2017.5029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N Engl J Med. Aug 20 2015;373(8):726–36. doi: 10.1056/NEJMoa1502309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Haroche J, Cohen-Aubart F, Emile JF, et al. Reproducible and sustained efficacy of targeted therapy with vemurafenib in patients with BRAF(V600E)-mutated Erdheim-Chester disease. J Clin Oncol. Feb 10 2015;33(5):411–8. doi: 10.1200/JCO.2014.57.1950 [DOI] [PubMed] [Google Scholar]
  • 109.Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAF. Nature. Mar 18 2010;464(7287):427–30. doi: 10.1038/nature08902 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Heritier S, Jehanne M, Leverger G, et al. Vemurafenib Use in an Infant for High-Risk Langerhans Cell Histiocytosis. JAMA Oncol. Sep 2015;1(6):836–8. doi: 10.1001/jamaoncol.2015.0736 [DOI] [PubMed] [Google Scholar]
  • 111.Cournoyer E, Ferrell J, Sharp S, et al. Dabrafenib and trametinib in Langerhans cell histiocytosis and other histiocytic disorders. Haematologica. Sep 21 2023;doi: 10.3324/haematol.2023.283295 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cohen Aubart F, Emile JF, Carrat F, et al. Targeted therapies in 54 patients with Erdheim-Chester disease, including follow-up after interruption (the LOVE study). Blood. Sep 14 2017;130(11):1377–1380. doi: 10.1182/blood-2017-03-771873 [DOI] [PubMed] [Google Scholar]
  • 113.Whitlock JA, Geoerger B, Dunkel IJ, et al. Dabrafenib, alone or in combination with trametinib, in BRAF V600-mutated pediatric Langerhans cell histiocytosis. Blood Adv. Aug 8 2023;7(15):3806–3815. doi: 10.1182/bloodadvances.2022008414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Tardieu M, Neron A, Duvert-Lehembre S, et al. Cutaneous adverse events in children treated with vemurafenib for refractory BRAF(V600E) mutated Langerhans cell histiocytosis. Pediatr Blood Cancer. Sep 2021;68(9):e29140. doi: 10.1002/pbc.29140 [DOI] [PubMed] [Google Scholar]
  • 115.Laetsch TW, DuBois SG, Bender JG, Macy ME, Moreno L. Opportunities and Challenges in Drug Development for Pediatric Cancers. Cancer Discov. Mar 2021;11(3):545–559. doi: 10.1158/2159-8290.CD-20-0779 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Milne P, Abhyankar H, Scull B, et al. Cellular distribution of mutations and association with disease risk in Langerhans cell histiocytosis without BRAFV600E. Blood Adv. Aug 23 2022;6(16):4901–4904. doi: 10.1182/bloodadvances.2022007519 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Allen CE, Li L, Peters TL, et al. Cell-specific gene expression in Langerhans cell histiocytosis lesions reveals a distinct profile compared with epidermal Langerhans cells. J Immunol. Apr 15 2010;184(8):4557–67. doi: 10.4049/jimmunol.0902336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Okamoto T, Murata Y, Hasegawa D, et al. Targeting of SIRPalpha as a potential therapy for Langerhans cell histiocytosis. Cancer Sci. May 2023;114(5):1871–1881. doi: 10.1111/cas.15758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sengal A, Velazquez J, Hahne M, et al. Overcoming T-cell exhaustion in LCH: PD-1 blockade and targeted MAPK inhibition are synergistic in a mouse model of LCH. Blood. Apr 1 2021;137(13):1777–1791. doi: 10.1182/blood.2020005867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Evseev D, Osipova D, Kalinina I, et al. Vemurafenib combined with cladribine and cytarabine results in durable remission of pediatric BRAF V600E-positive LCH. Blood Adv. Sep 26 2023;7(18):5246–5257. doi: 10.1182/bloodadvances.2022009067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Eckstein OS, Bernhardt MB, Hood CG, Karri V, Kralik SF, McClain KL. Treatment of Langerhans cell histiocytosis with subcutaneous cytarabine. Pediatr Hematol Oncol. 2023;40(5):497–505. doi: 10.1080/08880018.2022.2137609 [DOI] [PubMed] [Google Scholar]
  • 122.Morimoto A, Shioda Y, Imamura T, et al. Intensified and prolonged therapy comprising cytarabine, vincristine and prednisolone improves outcome in patients with multisystem Langerhans cell histiocytosis: results of the Japan Langerhans Cell Histiocytosis Study Group-02 Protocol Study. Int J Hematol. Jul 2016;104(1):99–109. doi: 10.1007/s12185-016-1993-3 [DOI] [PubMed] [Google Scholar]
  • 123.Morimoto A, Shioda Y, Imamura T, et al. Intensification of induction therapy and prolongation of maintenance therapy did not improve the outcome of pediatric Langerhans cell histiocytosis with single-system multifocal bone lesions: results of the Japan Langerhans Cell Histiocytosis Study Group-02 Protocol Study. Int J Hematol. Aug 2018;108(2):192–198. doi: 10.1007/s12185-018-2444-0 [DOI] [PubMed] [Google Scholar]
  • 124.Goyal G, Abeykoon JP, Hu M, et al. Single-agent cladribine as an effective front-line therapy for adults with Langerhans cell histiocytosis. Am J Hematol. May 1 2021;96(5):E146–E150. doi: 10.1002/ajh.26119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Diamond EL, Durham BH, Ulaner GA, et al. Efficacy of MEK inhibition in patients with histiocytic neoplasms. Nature. Mar 2019;567(7749):521–524. doi: 10.1038/s41586-019-1012-y [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES