ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Treatment of non-pulmonary Langerhans cell histiocytosis

Treatment of non-pulmonary Langerhans cell histiocytosis
Literature review current through: Jan 2024.
This topic last updated: Aug 19, 2022.

INTRODUCTION — Langerhans cell histiocytosis (LCH) is a neoplastic histiocytic disorder that most commonly affects bones and skin, but it can also involve bone marrow, liver, spleen, lungs, central nervous system, and other organs. LCH is rare, but it is considerably more common in children (especially younger children) than in adults.

Histiocytic disorders are categorized as LCH and non-Langerhans histiocytoses. The historical terms for LCH: histiocytosis-X, Letterer-Siwe disease, Hand-Schüller-Christian disease, and diffuse reticuloendotheliosis should be abandoned.

The BRAF V600E mutation is present in more than half of cases and activation of the mitogen-activated protein kinase (MAPK) pathway is a key driver of this neoplastic disorder [1-3].

Management of non-pulmonary LCH in children and adults is presented here.

Diagnosis and management of pulmonary LCH are discussed separately. (See "Pulmonary Langerhans cell histiocytosis".)

The epidemiology, clinical manifestations, pathologic features, diagnosis, and differential diagnosis of non-pulmonary LCH are presented separately. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

PRETREATMENT EVALUATION — Pretreatment evaluation of LCH should define the symptoms, involved organ systems, extent of their involvement, and effects on organ function.

Clinical manifestations, diagnosis, and differential diagnosis of LCH are detailed separately. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

Clinical – History and physical examination should define the type and severity of LCH-associated symptoms and signs.

Examples include constitutional symptoms (eg, fever, sweats, fatigue), pulmonary findings (eg, dyspnea, cough, hemoptysis, chest pain), musculoskeletal pain or myalgias, gastrointestinal symptoms (eg, diarrhea, melena, jaundice, abdominal fullness or pain), skin rash or subcutaneous nodules, lymphadenopathy, neurologic findings (eg, headache, ataxia, dysarthria, diplopia, altered hearing, cognitive decline, seizures), and endocrine symptoms (eg, polydipsia, polyuria, cold intolerance, growth delay, decreased libido).

Laboratory studies

Complete blood count (CBC), differential count, and reticulocyte count.

Coagulation – Prothrombin time (PT) and activated partial thromboplastin time (aPTT) for patients with hepatomegaly, jaundice, abnormal liver enzymes, or a low total protein or albumin.

Serum chemistries:

-Electrolytes, kidney function tests, calcium, liver function tests, total protein, albumin

-Thyroid stimulating hormone (TSH), free T4

-Prolactin and IGF-1

-Morning serum cortisol, ACTH, and osmolality

-Follicle stimulating hormone (FSH) and luteinizing hormone (LH) for males or estradiol for females

Morning urine osmolality.

Bone marrow examination should be performed in all children ≤2 years and in other patients if the CBC is abnormal or there are concerns for a myeloid neoplasm or hemophagocytic lymphohistiocytosis.

Pulmonary function tests for patients with respiratory findings or abnormal chest radiograph. (See "Pulmonary Langerhans cell histiocytosis", section on 'Pulmonary function testing'.)

Pathology – The characteristic microscopic appearance is of Langerhans cells ("coffee bean-shaped nucleus") amidst an inflammatory infiltrate of neutrophils, lymphocytes, macrophages, multinucleated giant cells, and eosinophils. Immunohistochemistry shows expression of CD1a and CD207 (langerin) and is discussed separately. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis", section on 'Pathologic features'.)

For all patients, we test the biopsy specimen for the BRAF V600E mutation using immunohistochemistry or polymerase chain reaction (PCR). If BRAF V600E is not detected, we test for mutations of the MAPK pathway (eg, ARAF, NRAS, KRAS, MAP2K1, PIK3CA) and fusion genes with a targeted tumor mutation panel.

Imaging:

Whole-body positron emission tomography (PET)/computed tomography (CT), including distal extremities, to determine the initial extent of organ involvement and as a baseline for assessing response to therapy [4,5].

For infants <1 year, we favor a bone scan to reduce radiation exposure.

For selected patients, based on clinical or laboratory evidence of organ involvement, the following may be obtained to stage the disease and monitor response to treatment:

-High-resolution chest CT for patients with respiratory symptoms or abnormalities on chest radiograph/PET-CT and for all children <1 year.

-Magnetic resonance imaging (MRI) of brain, pituitary gland, mastoids for patients with clinical or laboratory findings of arginine vasopressin deficiency (previously called central diabetes insipidus), neurologic abnormalities, or bony involvement of the head or neck.

-CT with contrast of abdomen and pelvis or ultrasound of liver and spleen.

-MRI of the spine for patients with vertebral involvement.

-Trans-thoracic echocardiogram (TTE) and right heart catheterization for selected patients with pulmonary LCH. (See "Pulmonary Langerhans cell histiocytosis".)

-Pulmonary function tests and exercise capacity testing for patients with respiratory symptoms or those with pulmonary LCH.

-Magnetic resonance cholangiopancreatography (MRCP) or endoscopic retrograde cholangiopancreatography (ERCP) for patients with cholestatic liver function tests (elevated alkaline phosphatase and/or bilirubin), or abnormal/dilated bile ducts on CT scan or ultrasound.

Other clinical testing – For selected patients, colonoscopy or abdominal ultrasound may be useful to monitor disease response when PET or other imaging cannot be used.

Prior to treatment with a targeted agent:

-Electrocardiogram (EKG) prior to BRAF-inhibitor treatment.

-TTE prior to MEK-inhibitor treatment.

OVERVIEW OF MANAGEMENT — Management of LCH is guided by the extent and severity of disease, as determined by the pretreatment evaluation. (See 'Pretreatment evaluation' above.)

Each of the following questions should be addressed for management of LCH [6]:

Central nervous system involvement – Is there involvement of the brain parenchyma, pituitary, or involvement of central nervous system (CNS)-"risk" bones (see below)? (See 'Multiple bone lesions' below.)

Involvement of the CNS requires systemic therapy because it is associated with adverse prognosis and increased risk of neurologic damage. The classic "high risk" organs in patients with LCH are the liver, spleen, and bone marrow, as their involvement confers a higher risk of treatment failure and death [7,8]. Importantly, management of critical/risk organ involvement differs according to the patient's age (ie, ≤20 years versus >20 years). (See 'Children' below and 'Adults' below.)

Single-system versus multisystem involvement – Is there involvement of only a single organ system (eg, bone, skin) versus involvement of more than one organ system?

Single system – Single bone lesion LCH can often be managed with local treatment (curettage), while limited skin involvement may be managed with local or systemic therapy. Management of single-system disease is similar for children and adults, except where specifically noted. (See 'Single system (children and adults)' below.)

Note that evaluation and management of isolated pulmonary LCH (ie, involvement of lungs, alone) are discussed separately. (See "Pulmonary Langerhans cell histiocytosis".)

Multisystem – Multisystem LCH typically requires systemic therapy; management of multisystem LCH differs for children and adults, as discussed below. (See 'Multisystem' below.)

Unifocal versus multifocal involvement – Within a single organ system, is the involvement limited to a single site (unifocal; eg, a single bone lesion), or is it more widespread (multifocal; eg, extensive skin involvement)?

Management of unifocal versus multifocal involvement is discussed within the affected organ system (eg, bone, skin). (See 'Bone' below and 'Skin' below.)

Symptoms – Does the patient have troublesome symptoms or require treatment to prevent progression or complications of LCH or is the disease asymptomatic?

Our approach to management of LCH is consistent with guidelines from the Histiocytosis Association of America and the Euro-Histio-Net [6]. We encourage participation of both children and adults in a clinical trial, when possible. The website for the Histiocytosis Association of America and an international directory of affiliated organizations can be found at histio.org, histiocytesociety.org, or clinicaltrials.gov.

SINGLE SYSTEM (CHILDREN AND ADULTS) — For both children and adults with single-system LCH, treatment is guided by the involved organ and the extent of disease.

Treatment for single-system LCH is similar for children and adults, except as otherwise noted.

Bone — Management of bone disease is stratified as follows:

Central nervous system (CNS) risk bones – Involvement of the mandible, orbit, mastoid, temporal, or sphenoid bones is associated with increased risk for CNS involvement; this association is better documented in children than in adults. Nevertheless, treatment requires systemic therapy because these sites are challenging to treat safely and effectively with surgery and/or radiation therapy (RT). Note that children should not be treated with RT.

Systemic management for CNS risk bones differs for children versus adults, as described below. (See 'Multisystem' below.)

Non-CNS risk bone lesions:

Single lesion – Management of a single, non-CNS risk bone lesion is the same for children and adults, as described below. (See 'Single bone lesion' below.)

Multiple bone lesions – Management of other bone disease, including two or more lesions, a single lesion ≥5 cm, and femoral or vertebral involvement requires systemic therapy, as described below. (See 'Multisystem' below.)

Single bone lesion — For a single bone lesion, we generally offer curettage, which provides both a tissue diagnosis and adequate treatment for most patients. RT is a reasonable alternative for selected cases or to enhance symptom control, but it is rarely administered to children. No prospective studies have directly compared curettage versus RT. Spontaneous regression of solitary bone lesions has been reported [9,10].

Note that a single bone lesion ≥5 cm or involvement of a femur, vertebra, or CNS-risk bone (ie, orbit, mastoid, temporal, or sphenoid bone) is managed with systemic therapy, as described below. (See 'Systemic therapy' below.)

Options for the management of a single non-CNS risk bone lesion are:

Biopsy/curettage – Curettage of a single bone lesion is suitable to establish the diagnosis of LCH, initiate healing, and enable adjacent normal bone to regrow.

Complete excision (ie, "clean margin") of bone lesions is not necessary; complete excision may increase the size of the bony defect, prolong the time for healing, and/or cause permanent skeletal defects. Intra-lesional injection of a glucocorticoid (eg, methylprednisolone 125 mg) may hasten healing and lessen pain at the biopsy site. Clinical judgment should be used to determine if bone grafting is needed.

Curettage is associated with a low recurrence rate (eg, 4 to 10 percent) for small bony lesions [11,12]. Treatment of bone LCH is similar in children and adults, but curettage may be less effective and spontaneous remissions less common in adults. One study compared recurrence rates between skeletally-mature and skeletally-immature patients with solitary LCH [13]. Among 17 skeletally-immature children who underwent biopsy with observation alone or curettage there were no recurrences beyond three years; by contrast, one-quarter of skeletally-mature patients had recurrence following biopsy, curettage, and bone grafting.

Radiation therapy – RT can be helpful for persistent or recurrent lesions in adults after curettage or systemic therapy. RT should not be used in children.

RT (eg, 6 to 12 gray [Gy] delivered at 2 Gy per fraction) has been used in teenagers and adults with an impending neurologic deficit or if the surgical risk is high (eg, a lesion of the cranial base or odontoid peg) [6,14]. However, we generally favor treatment with single-agent chemotherapy (eg, cytarabine or cladribine), rather than RT for children or adults with osseous sites that are unsafe for RT. (See 'Cytarabine' below and 'Cladribine' below.)

A survey of 30 adults with bone LCH who were treated with various combinations of surgery, radiotherapy, and corticosteroids reported recurrence in one-third, with the lowest rate of recurrence in those who had surgery and RT [15].

Bisphosphonate therapyPamidronate or zoledronic acid may lessen symptoms or improve healing for patients with one or two bone lesions.

Anecdotal reports and small series describe reduced pain, improved function, and/or radiographic response in most patients with osteolytic lesions from LCH when a bisphosphonate was added to surgery or RT in children or adults [16-19]. In one study, responses were seen in 12 of 16 children treated with six courses of pamidronate at four-week intervals, but the role of pamidronate in the healing of bone and other lesions is uncertain [20].

Multiple bone lesions — For patients with two or more bone lesions, a single bone lesion ≥5 cm, femoral or vertebral involvement, or involvement of a CNS risk bone (ie, orbit, mastoid, temporal, or sphenoid bones), treatment involves systemic therapy. Adjunctive surgical techniques (and/or RT for adults) may be added to lessen the risk of complications.

Note that the preferred systemic treatment differs for children versus adults:

Systemic therapy

Children – For patients ≤20 years with multiple bone lesions, we treat with systemic therapy, as described below. (See 'Children' below.)

Adults – For adults with multiple bone lesions who do not have CNS or risk organ involvement, we treat with systemic therapy, as described below. (See 'No CNS or risk organ involvement' below.)

Adjunctive treatment — Surgical stabilization, bone grafting, and/or RT may be used to prevent complications with weight-bearing bones. Involvement of the spine or femora can cause bone pain, fracture from weight-bearing, or vertebral collapse/spinal cord compression.

Surgery – Orthopedic intervention with bracing or bone grafting may be necessary for patients with neurologic symptoms or unstable lesions. For children with lesions of the spine (eg, vertebra plana), bed rest and external immobilization with a rigid orthesis may enable reconstitution of vertebral body height because this does not involve the endochondral ossification centers [21].

Radiation – RT should be considered for adult patients with bone lesions of vertebrae or the femoral neck, which are at risk of collapse, but the benefit must be weighed against adverse effects, including potential risk of damage to endochondral ossification centers in children [22,23].

RT for LCH offers symptomatic and radiographic responses in adults; it has only rarely been used in children [24]. LCH-related bone pain generally resolves within four months and provides long-term control for most patients.

Skin — It is important to exclude involvement of other organs in patients who appear to have skin-only LCH, as many children who present with cutaneous involvement also have multisystem LCH [25]. Clinical evaluation, laboratory studies, and PET-CT to evaluate systemic disease is discussed above. (See 'Pretreatment evaluation' above.)

Patients with skin-only disease may have spontaneous regression of cutaneous LCH (ie, "self-healing cutaneous LCH") or they may progress to multisystem involvement; up to 40 percent of infants have been reported to progress to multisystem involvement [26-28]. There are no prognostic factors or biomarkers to confidently predict the course of skin-only disease and the natural history only becomes apparent with clinical investigation and follow-up.

Management of skin disease is informed by the presence of symptoms and the extent of involvement (eg, unifocal versus multifocal or extensive disease):

Asymptomatic – Observation alone is acceptable for asymptomatic patients with limited (eg, small surface area) LCH, if that is acceptable to the patient and/or caregivers. Some patients may favor treatment as described for symptomatic disease.

Symptomatic and/or extensive skin disease – We treat symptomatic and/or extensive skin involvement with topical therapy or an oral agent. The choice of treatment varies with the severity and extent of disease and patient preference.

Topical therapy

-Steroid – Topical triamcinolone

-Nitrogen mustard (mechlorethamine) 20 percent

Methotrexate 20 mg weekly by mouth, as a single agent or in combination with [29-31]:

-Prednisolone (20 to 40 mg/m2 twice daily)

-6-mercaptopurine (50 mg/m2 daily)

-Hydroxyurea (10 mg/kg twice daily)

Lenalidomide 25 mg on days 1 to 21 in 28-day cycles [32]. Thromboprophylaxis should be considered when administering lenalidomide or thalidomide. (See "Multiple myeloma: Prevention of venous thromboembolism", section on 'Choice of VTE prophylaxis'.)

Thalidomide 100 mg daily, by mouth [33].

Small retrospective studies and case reports have described responses to topical steroids, topical nitrogen mustard, and oral methotrexate, hydroxyurea, lenalidomide, and thalidomide [29,31-36], but there are no prospective comparisons of these agents for cutaneous LCH. The combination of methotrexate plus hydroxyurea has been particularly successful for women with genital lesions [31].

Other non-critical organs — Lymph nodes, thyroid, and thymus are considered non-critical organs, but they require systemic chemotherapy, as described for multisystem involvement. (See 'Multisystem' below.)

Lymph nodes that drain another LCH lesion are not considered lymph node involvement. In addition, it is important to rule out reactive infiltration of a lymph node by non-neoplastic Langerhans cells [37].

Spontaneous regression of lymph node involvement has been reported. Extensive surgery (eg, neck dissection) should not be performed to avoid the adverse surgical effects.

Management of specific sites such as oral cavity and gingiva may include hydroxyurea or oral methotrexate.

For a single polyp in the gastrointestinal tract, observation or endoscopic excision are acceptable. For more extensive gastrointestinal involvement, systemic therapy should be offered.

Evaluation, diagnosis, and management of lung involvement (isolated pulmonary LCH) are discussed separately. (See "Pulmonary Langerhans cell histiocytosis".)

MULTISYSTEM — Management of multisystem LCH is risk-adjusted and based on the patient's age.

The level of risk is determined by whether there is involvement of the central nervous system (CNS) and/or one of the critical ("risk") organs (ie, bone marrow, liver, or spleen).

High risk High-risk multisystem LCH includes involvement of CNS and/or a risk organ.

Children – Management of high-risk multisystem LCH in children (<20 years old) is described below. (See 'Vinblastine-prednisone induction therapy' below.)

Adults – Management of high-risk multisystem LCH in adults is described below. (See 'CNS or risk organ involvement' below.)

Low-risk – Low-risk multisystem LCH may involve skin, bone, lymph nodes, thymus, hypophysis, and gastrointestinal tract, but not the CNS or a risk organ.

Children – Management of low-risk multisystem LCH in children (<20 years old) is described below. (See 'Vinblastine-prednisone induction therapy' below.)

Adults – Management of low-risk multisystem LCH in adults is described below. (See 'No CNS or risk organ involvement' below.)

Children — For initial treatment of children with multisystem LCH, we suggest induction therapy with vinblastine plus prednisone (V-P), rather than other chemotherapy regimens or a targeted agent. This suggestion is based on high rates of response and tolerable toxicity with V-P; by contrast, there is limited experience with targeted agents for initial treatment in children.

V-P induction therapy is associated with an objective response in approximately 90 percent of children, including nearly two-thirds of children with critical organ involvement; V-P is generally well-tolerated in children, but half of patients relapse or progress [8,38]. Details of V-P therapy in children are presented in Outcomes (below).

Vinblastine-prednisone induction therapy — V-P is efficacious and well-tolerated in children and is considered the standard systemic treatment for patients ≤20 years old [8]. Subsequent management is guided by the response to induction therapy. (See 'Continuation phase' below.)

Administration – We treat both high-risk (CNS and/or risk organ involvement) and low-risk patients with the following induction therapy:

Vinblastine (6 mg/m2 intravenous bolus weekly) for six weeks

Prednisone (40 mg/m2 daily by mouth for four weeks, then tapering over two weeks)

Response should be assessed within two weeks of completing induction therapy, as described below. (See 'Response assessment' below.)

Subsequent treatment is guided by the response to induction therapy and whether there was CNS or risk-organ involvement at the time of induction initiation, as described below. (See 'Continuation phase' below.)

Adverse effects – The most common side effects of vinblastine are constipation, mild cytopenias, and, rarely, alopecia. Peripheral neuropathy is more common in teenagers than in younger children. Over the course of 12 months therapy, prednisone causes excessive appetite and weight gain and mood changes in many; it may be associated with hypertension, glucose intolerance, osteopenia, and myalgias. (See "Overview of neurologic complications of conventional non-platinum cancer chemotherapy", section on 'Other vinca alkaloids'.)

Outcomes – V-P is effective and well-tolerated in children, but it has limited efficacy and is poorly tolerated in patients >20 years.

In the LCH-III study, >500 children (<18 years) were stratified for treatment as high-risk (CNS or risk-organ involvement) versus low-risk (no CNS or risk-organ involvement) [8].

High-risk patients were randomly assigned to induction with V-P versus V-P plus methotrexate, followed by 12 months of continuation therapy (using the induction regimen plus mercaptopurine). Addition of methotrexate did not increase the response rate, accelerate disease resolution, or increase survival, but it was associated with increased toxicity. Outcomes included 71 percent response, 84 percent five-year overall survival (OS), and 27 percent relapse/progression. Grade ≥3 toxicity (eg, infections, cytopenias, hepatotoxicity) was more common in the patients treated with methotrexate (30 versus 7 percent).

Low-risk patients received V-P induction and responders were randomly assigned to 6 versus 12 months of continuation therapy. Responses occurred in 86 percent, but longer continuation therapy was associated with lower reactivation rates at three years (34 versus 44 percent, respectively) and at five years (37 versus 54 percent). Toxicity was mild in both arms.

Results in LCH-III were superior to outcomes from earlier studies [38]. LCH-II used the same induction regimen but included only six months of continuation therapy [39], while LCH-I compared induction with a glucocorticoid plus vinblastine versus a glucocorticoid plus etoposide [40]. In LCH-III, five-year OS in high-risk patients (84 percent) was superior to that in LCH-II (69 percent) and LCH-I (62 percent); five-year reactivation was also lower in LCH-III compared with LCH-II and LCH-I (27 versus 44 and 55 percent, respectively). Other modifications of the V-P regimen (eg, increased prednisone dose, adding cytarabine) and lengthening continuation therapy did not substantially increase the response rate or improve event-free survival (EFS) among 82 children [41].

Ongoing studies are testing if addition of mercaptopurine to V-P is beneficial for high-risk patients (LCH-IV) and comparing V-P versus single-agent cytarabine (NCT02670707). We encourage participation in a clinical trial, when possible. The website for the Histiocytosis Association of America and an international directory of affiliated organizations can be found at histio.org, histiocytesociety.org, or clinicaltrials.gov.

Importantly, V-P is generally neither efficacious nor well-tolerated in patients >20 years, as described below. (See 'Adults' below.)

Continuation phase — Relapse can be expected if no continuation therapy is given after achieving a response with V-P induction therapy [40]. Continuation therapy is stratified according to involvement of critical organs at initiation of induction therapy and the extent of response:

Complete response (CR) after six-week induction treatment

Low-risk – Continuation therapy comprises vinblastine (6 mg/m2 intravenously once every three weeks) plus prednisone (40 mg/m2 daily in three doses orally on days 1 to 5 every three weeks) for a total therapy time of 12 months.

High-risk – For children who presented with CNS or risk-organ involvement, continuation therapy adds mercaptopurine (50 mg/m2 daily orally) to prednisone (40 mg/m2 daily in three doses orally on days 1 to 5) and vinblastine (6 mg/m2 intravenously per day once every three weeks) for 12 months.

Partial response – Repeat V-P induction therapy. (See 'Vinblastine-prednisone induction therapy' above.)

Stable disease or progressive disease – Treat with a second-line chemotherapy regimen. (See 'Relapsed or refractory disease' below.)

After completing a year of treatment, we reassess patients clinically every three months for two years, every six months for two years, and then yearly. Repeat imaging is done at 3, 6, and 12 months and then stopped, unless clinical symptoms or signs indicate the need for repeat imaging.

Adults — We stratify treatment for multisystem LCH in adults according to whether the CNS and/or one of the "risk" organs (ie, bone marrow, liver, spleen) is involved.

No CNS or risk organ involvement — For adults with multisystem LCH that does not involve the CNS or a risk organ, we suggest single-agent cytarabine or cladribine, rather than combination chemotherapy or a targeted agent. Both single agents had superior efficacy and less toxicity compared with V-P in a study that included 58 adults with bone disease or multisystem LCH [42]; they have not been directly compared with a targeted agent.

Some patients with BRAF V600E-mutated multisystem LCH may warrant treatment with a BRAF inhibitor, rather than chemotherapy; advantages and disadvantages of these approaches are discussed below. (See 'BRAF V600E LCH' below.)

No prospective studies have directly compared cytarabine versus cladribine, and the preferred agent varies between institutions. While six or more cycles of cladribine therapy can be associated with prolonged thrombocytopenia and lymphopenia, two to four monthly cycles are effective and unlikely to cause severe cytopenias and/or infections [43]. Some experts favor cytarabine for patients with multifocal bone disease and favor cladribine for brain involvement [42,44,45]. Single-agent chemotherapy options are discussed below. (See 'Cytarabine' below and 'Cladribine' below.)

We encourage participation in a clinical trial, when possible. The website for the Histiocytosis Association of America and an international directory of affiliated organizations can be found at histio.org, histiocytesociety.org, or clinicaltrials.gov.

Cytarabine — Low dose cytarabine can be effective for patients with multisystem LCH, including multiple bone lesions, but it is generally not administered for patients who have extensive skin lesions.

AdministrationCytarabine 100 mg/m2 over 30 minutes daily for five days, repeated monthly for 12 months.

Toxicity – Most toxicity is hematologic; grade ≥3 adverse events (AE) were reported in 20 percent of adults in one report [42]. Some patients develop high fevers and/or hypotension with cytarabine.

Outcomes – A retrospective study reported that cytarabine achieved and sustained a response for one year in 79 percent of 24 adults; most patients had bone involvement, while 43 percent had other sites of disease [42]. In this study, cytarabine was more effective and less toxic than V-P. Cytarabine was also more efficacious than cladribine (79 versus 41 percent responses) and less toxic (grade ≥3 AEs 20 percent versus 37 percent), but cladribine toxicity reflected longer treatment (six cycles) than current practice (described below).

Cladribine — Treatment with single-agent cladribine (2-chlorodeoxyadenosine [2-CdA]) is well-tolerated and efficacious for adults and children with LCH.

AdministrationCladribine is administered as 5 mg/m2 (or 0.14 mg/kg) intravenous daily for five days, repeated monthly. We generally treat with two cycles of cladribine, followed by disease restaging. If there is no response, we discontinue cladribine and switch to alternative treatment (targeted therapy or cytarabine). If there is at least a partial response (PR) to two cycles, we administer two more cycles. We typically use four cycles, but no more than six cycles. Some patients with a PR continue to respond without further therapy [43]. Prophylaxis is generally given for Pneumocystis jirovecii pneumonia, varicella-zoster, and herpes simplex virus, as these patients become profoundly lymphopenic.

ToxicityCladribine is associated with lymphopenia, thrombocytopenia, and febrile neutropenia. Cytopenias can be prolonged, especially after >4 cycles of therapy.

Outcomes:

Adults – Single-agent cladribine was associated with 79 percent response rate (including 26 percent CR) in a retrospective study that included 38 adults (median 45 years) with multifocal LCH (82 percent with multisystem LCH) [44]. Cladribine was the front-line treatment for three-quarters of the patients and responses were reported in bone, lymph node, skin, lung, and pituitary; a median of four cycles of cladribine was administered. Grade ≥3 AEs included two patients with leukopenia that required treatment delay and one patient with febrile neutropenia; one patient died from sclerosing cholangitis, which is a rare complication of LCH. Five-year progression-free survival (PFS) and OS were 58 percent and 75 percent, respectively.

In another retrospective study, cladribine was less efficacious and associated with more AEs (75 percent grade ≥3 neuropathy) than cytarabine among adults with bone-predominant disease [42].

Children – Treatment with cladribine (median six cycles) in 44 children (median age four years; none with risk-organ involvement) was associated with 57 percent objective response, 14 percent stable disease, and 30 percent progressive disease [46]. Five-year OS was 98 percent and five-year cumulative incidence of disease progression or reactivation was 34 percent. Lymphopenia <500/microL was reported in 72 percent; grade ≥3 AEs were uncommon and there was no neuropathy or second malignancies.

In another cooperative group study (LCH-S-98), cladribine was associated with 75 percent response rate in children with low-risk LCH [47].

Combination chemotherapy in adults — Combination chemotherapy is generally not administered to adults (ie, >20 years) because it was shown to be more toxic and less efficacious than single-agent cytarabine or cladribine, as discussed above. (See 'No CNS or risk organ involvement' above.)

Vinblastine plus prednisone – Treatment of 19 adults with V-P (the standard combination chemotherapy for children) was associated with grade ≥3 neuropathy in 75 percent but achieved and sustained a response for one year in only 16 percent of patients [42]. Neuropathy caused by V-P can be irreversible for some patients [48]. A retrospective multicenter study that included 35 adults (80 percent with multisystem LCH) reported 70 percent response to V-P, no grade ≥3 peripheral neuropathy, but 40 percent of patients experienced LCH reactivation [49].

Other combination chemotherapy regimens that have been evaluated for LCH in adults include:

Vindesine plus prednisone – A retrospective single-center study reported that vindesine plus prednisone was associated with 64 percent response, which was comparable to the 70 percent response with more intensive CEVP (cyclophosphamide, etoposide, vindesine, prednisone) [50]. However, vindesine-prednisone in adults was associated with more disease recurrence and deaths compared with pediatric patients [51].

Methotrexate plus cytarabine – Treatment of adults with cytarabine plus methotrexate (MTX) was associated with high rates of response but substantial toxicity. Six cycles of MTX (1 g/m2 body surface area [BSA] intravenously) on day 1 and cytarabine (0.1 g/m2 BSA intravenously) on days 1 to 5 of every 28-day cycle was associated with 100 percent response (17 percent CR), 97 percent OS, and 49 month estimated EFS, with median follow-up of 44 months [52]. However, grade ≥3 neutropenia and thrombocytopenia were reported in 78 and 61 percent, respectively. Another study reported 88 percent response rate, but grade ≥3 cytopenias in 94 percent and 48 percent febrile neutropenia [53].

MACOP-B – A retrospective report of seven patients treated with MACOP-B (cyclophosphamide, doxorubicin, vincristine, methotrexate, bleomycin, and prednisone) reported five CR and two PR; three patients relapsed from 5 to 62 months after treatment [54]. No patient required discontinuation of treatment for toxicity and only two patients had a treatment delay due to grade 3 neutropenia.

CNS or risk organ involvement — Involvement of the CNS and/or a critical/risk organ (ie, bone marrow, liver, or spleen) is associated with inferior outcomes.

BRAF V600E LCH — For adults with BRAF V600E-mutated LCH who have CNS involvement or involvement of a critical/risk organ (ie, bone marrow, liver, or spleen) who are systemically ill, we suggest treatment with a BRAF inhibitor (eg, vemurafenib, dabrafenib), rather than systemic chemotherapy. BRAF inhibitors act quickly to prevent progression in nearly all patients and are associated with acceptable toxicity, but they require prolonged therapy; BRAF inhibitors have not been compared with chemotherapy in a prospective trial.

Some experts also favor a BRAF inhibitor for treatment of patients with BRAF V600E-mutated LCH who have ascending cholangitis or neurodegenerative LCH, which are rare complications of LCH. However, there is only limited, primarily anecdotal experience in these settings. Treatment with vemurafenib or dabrafenib resulted in clinical and magnetic resonance imaging (MRI) improvements in three of four patients [55]. Other aspects of management of neurodegenerative LCH and ascending cholangitis are discussed below. (See 'Non-BRAF V600E LCH' below.)

There is no evidence that either of the BRAF inhibitors is more effective or better tolerated than another. Patients with intolerance for one BRAF inhibitor may be able to switch to an alternate BRAF inhibitor without the same adverse effects. Follow-up with BRAF inhibitors has been limited and they can cause cutaneous toxicity (including cancers), are expensive, have limited availability, and may require life-long treatment. (See "Systemic treatment of metastatic melanoma with BRAF and other molecular alterations", section on 'BRAF V600 mutant disease'.)

Some patients or clinicians may favor chemotherapy, instead of a targeted agent. Advantages and disadvantages of chemotherapy versus a targeted agent include:

Single-agent chemotherapy – Treatment with chemotherapy requires frequent medical visits for intravenous administration and is associated with hematologic and other AEs. However, chemotherapy is only administered for a limited period (eg, six months) and it may achieve long-term disease control and potential cure. Many patients favor chemotherapy to avoid the life-long treatment that may be needed for a targeted agent.

BRAF inhibitors – Targeted agents are administered orally, but they require prolonged therapy; some targeted agents are associated with development of secondary cancers, while discontinuation of BRAF inhibitors may lead to recurrence of disease. Agents that target mutant BRAF (BRAF is mutated in more than half of LCH cases) are associated with prompt, robust, and potentially long-lasting responses. In practice, patients who have achieved a good response can be maintained in remission with chronic low-level dosing, with planned treatment breaks and close monitoring.

Vemurafenib has demonstrated efficacy in patients with LCH whose tumors carry the BRAF V600E mutation [56]. The VE-BASKET trial, which included four patients with LCH, reported positron emission tomography (PET) complete metabolic response in all cases [57]. Treatment of six adults with vemurafenib or dabrafenib was associated with a response in 83 percent (33 percent CR, 50 percent PR), median time to response was four to six months, and three patients with CNS disease attained a sustained remission >12 months [58]. Cutaneous AEs are common, including squamous cell carcinomas (SCC) and keratoacanthomas; one patient developed sclerosing cholangitis while on vemurafenib resulting in death.

Non-BRAF V600E LCH — For adults with CNS or risk-organ involvement whose tumor does not harbor a BRAF V600E mutation, we suggest treatment with single-agent cytarabine or cladribine, rather than combination chemotherapy or a targeted agent. Initial treatment using targeted agents other than BRAF inhibitors is encouraging, but there is only limited published experience with them in LCH, as described below. Single-agent chemotherapy is more efficacious and less toxic than combination chemotherapy in adults.

Administration, toxicity, and outcomes with cytarabine and cladribine are presented above. (See 'Cytarabine' above and 'Cladribine' above.)

There are special considerations for treatment of the following conditions:

CNS involvement

Mass lesions – We favor cladribine for mass lesions of the brain and pituitary. If cytarabine is given, we treat with a higher dose (eg, 150mg/m2) and/or in combination with methotrexate.

Chemotherapy can be combined with resection and/or radiation therapy (RT), according to clinical judgment, but multimodal approaches have not been evaluated in a prospective trial.

Mass lesions of the white or grey matter, hypothalamus, and pituitary have been effectively treated with clofarabine, cladribine, or V-P [45,48,59]. Rarely, resection of a solitary brain mass or enlarged pituitary is done, when occurring in a patient with known LCH. Low dose RT has achieved remission, according to a case series [60]. Treatment of the pituitary damaged by LCH did not reverse arginine vasopressin deficiency, although some patients required lower doses of antidiuretic hormone (desmopressin [DDAVP]) after receiving chemotherapy [61].

Neurodegenerative LCH – We generally treat this rare complication of LCH with cladribine or with an increased dose of cytarabine (eg, 175 mg/m2), based on our clinical experience. The role of targeted therapy is still evolving in this setting and may be a reasonable alternative [62].

Cytarabine, with or without vincristine, was reported to provide improvement in clinical (six of eight patients) and MRI findings (five of eight patients) [63]. All but two of seven patients had stable neurologic and radiographic findings for >10 years after stopping therapy.

Ascending cholangitis – This grave complication is poorly responsive to chemotherapy and patients should be promptly referred for consideration of liver transplantation.

There is very limited published experience with initial treatment of LCH with targeted agents against mutations of MEK1, MEK2, or CSF1R and rearrangements in RET and ALK kinases [64,65]. However, we have used these agents in selected cases with robust responses that are similar to those with BRAF-inhibitors. We begin treatment with cobimetinib (MEK inhibitor) using a reduced dose (eg, 40 mg daily for 21 days, 28-day cycles) and monitor the patient for retinopathy, reduced cardiac output, and cutaneous toxicity, including skin cancers. (See "Systemic treatment of metastatic melanoma with BRAF and other molecular alterations", section on 'Dosing considerations and toxicities of BRAF plus MEK inhibitors'.)

Cobimetinib was associated with 90 percent response rate and no relapses in a study of 22 patients with various histiocytic disorders, including two with LCH and two with mixed histiocytosis [65]. Responses were seen in all cases, regardless of presence of a MAPK-pathway mutations. In patients who are critically ill and mutational status is unknown, empiric MEK-inhibitor therapy is a reasonable approach. As mentioned for BRAF-inhibitors above, MEK-inhibitors and other targeted agents likely require a prolonged duration of therapy, due to the risk of disease relapse at discontinuation.

Cobimetinib is approved by the US Food and Drug Administration for use as a single agent for the treatment of adult patients with histiocytic neoplasms.

RESPONSE ASSESSMENT — We assess response to treatment for LCH using 18-fluorodeoxyglucose (FDG) positron emission tomography (PET). PET is sensitive for detection of most LCH and it typically demonstrates a response before it is apparent with other modalities (eg, bone scan, plain radiography) [5].

For settings where PET is not useful (eg, brain lesions), when there are no imaging abnormalities (eg, gastrointestinal [GI] tract), or if PET is not available or contraindicated (eg, infants), we use clinical evaluation (including endoscopy for GI lesions), computed tomography (CT), and/or magnetic resonance imaging (MRI) to assess response. Circulating BRAF-mutated cells can be monitored in the peripheral blood by a sensitive polymerase chain reaction (PCR) test, if available [66].

Timing of response assessment depends on the sites of disease:

Single-system disease – We generally review symptoms, examine involved sites, and repeat PET/CT two months after curettage of a bone lesion or treatment of skin disease.

Multisystem disease

Children – PET/CT imaging should be repeated after completion of induction therapy.

Adults – We assess response to chemotherapy after two cycles of chemotherapy or two months of a targeted agent, or as clinically indicated.

Responses are defined as:

Complete response (CR):

PET – Normalization of lesions with FDG uptake equal to surrounding background tissue.

CT or MRI – Complete anatomic resolution of lesions or abnormal features (eg, enhancement, diffusion restriction).

Partial response (PR):

PET – Reduction from baseline standardized uptake value (SUV) of lesions, but persistent uptake greater than surrounding background tissue.

CT or MRI – Reduction, but not complete resolution of lesions or abnormal imaging features.

Progressive disease (PD):

PET – Appearance of new FDG-avid lesions or increased SUV value of previously detected lesions.

CT or MRI – Appearance of new lesions or worsening of abnormal features or growth of existing lesions.

Stable disease (SD): Does not meet any of the criteria described above.

For patients with BRAF V600E-mutated LCH, the level of circulating tumor (ct) BRAF V600E DNA has been reported to correlate with high-risk clinical characteristics, higher risk of relapse, and more extensive disease [1,67-71]. However, we await further validation before using this clinically, because apparent responses may reflect low levels of circulating cells.

RELAPSED OR REFRACTORY DISEASE — Management of relapsed or refractory (r-r) LCH should be individualized according to the timing of relapse, sites of disease, performance status, and previous treatment. Only case reports and small case series are available to guide the care of these patients; we encourage enrollment in a clinical trial, when available.

Single-system disease – Management of r-r single-system disease can use alternate localized therapies or systemic therapy, as described for multisystem disease.

Bone-only – For bone disease that recurs after curettage, treatments may include radiation therapy, chemotherapy (cytarabine or cladribine), and/or a bisphosphonate. (See 'Bone' above.)

Skin-only – For recurrence after topical therapy, treatment with an alternate approach, such as an oral agent (eg, hydroxyurea, methotrexate, lenalidomide, or thalidomide) can provide disease control and symptomatic relief [30-33]. (See 'Skin' above.)

Multisystem disease – For r-r multisystem LCH, we generally offer systemic treatment that differs from the initial therapy.

Children – For children who relapse >12 months after completing vinblastine plus prednisone (V-P), retreatment with V-P alone or combined with oral methotrexate or mercaptopurine can be effective; approximately 85 percent of patients achieved a second remission using this approach [72].

For other cases of r-r LCH in children, we generally treat with single-agent cytarabine or cladribine. If a BRAF mutation is present, a BRAF inhibitor may be offered.

Outcomes with treatment of r-r LCH in children include:

-Cladribine – Response to cladribine depended on involvement of critical organs (62 percent response for no risk-organ involvement versus 22 percent response with risk-organ involvement) [47]. A retrospective analysis reported responses to cladribine in 4 of 10 patients with refractory high-risk disease [73].

-CytarabineCytarabine has been used as a single agent [74]. Reduced doses of cladribine and cytarabine together with other agents (eg, vincristine, mercaptopurine, and others) was active for children with relapsed LCH [75].

-ClofarabineClofarabine has demonstrated efficacy and good tolerability in children with refractory LCH (90 percent survival) [76-78]. Our experience is that this is an especially effective therapy for patients with refractory LCH of bones, skin, lymph nodes, and liver.

-Cladribine plus high-dose cytarabine – Higher-dose cladribine plus high-dose cytarabine has been used for children with relapsed or refractory high-risk LCH [79-81]. Treatment of 27 patients with refractory, risk-organ-positive LCH reported 92 percent response rate and 85 percent (95% CI 65-94 percent) estimated five-year overall survival (OS) [81]. Long periods of pancytopenia may occur after each course.

-Targeted agents – BRAF inhibitors can achieve a response in patients with neurodegenerative LCH, but progression-free survival (PFS) was only 33 percent [71]. A study of 54 children reported 100 response after two months of therapy, but PFS was approximately 5 percent [69]. BRAF inhibitors may be particularly effective in children with bone marrow and liver LCH that were refractory to standard therapy. Dabrafenib combined with trametinib (MEK inhibitor) achieved a rapid and sustained response in a patient with BRAF V600E-mutant LCH [68]. Treatment with BRAF inhibitors is discussed above. (See 'BRAF V600E LCH' above.)

Adults – For adults who relapse after treatment with cytarabine or cladribine, we generally treat with a mutation-directed targeted agent or an alternative chemotherapy agent. The choice of therapy should be individualized based on patient and disease-specific factors.

-Chemotherapy – There is limited published experience with switching from one chemotherapy regimen to another in relapsed LCH. For patients who relapse after V-P-based therapy, single-agent cytarabine or cladribine is a good choice. Switching treatments from cladribine to cytarabine (and vice versa) can be considered if the relapse occurs >1 year after initial treatment.

-Targeted agents:

BRAF inhibitors – Three patients with BRAF-mutated combined LCH/Erdheim-Chester disease had sustained responses to vemurafenib after failing other treatments [56]. In the VE-Basket study, four LCH patients and 22 Erdheim-Chester patients were treated with vemurafenib as salvage therapy [57]. Three of the four patients with LCH had complete responses; adverse effects included arthralgias, rashes, fatigue, and hypertension. Given the efficacy reported in these and other series, BRAF-inhibitors are now being used as front-line therapy [58]. (See 'BRAF V600E LCH' above.)

Inhibitors of MEK and other mutations – There is less published information about the role of other targeted therapies for relapsed LCH. The MEK inhibitor, cobimetinib, was associated with responses in patients with or without the BRAF V600E mutation, but most patients required dose reduction for toxicity (eg, decreased cardiac ejection fraction, acneiform rash, diarrhea, retinopathy) [65]. Other reports include treatment of r-r LCH with vemurafenib, dabrafenib, trametinib, cobimetinib [55,71,82-86]. The tyrosine kinase inhibitor, imatinib, was associated with responses in three adults (with skin, lung, bone, and/or central nervous system involvement) [87,88], but not in others [89].

Targeted agents for genes other than BRAF V600E have been used for treatment of other histiocytic or hematologic malignancies, but their utility for LCH is unproven. Examples include crizotinib (ALK gene fusion) [64], selpercatinib (RET gene fusion), pexidartinib (CSF1R mutation), and entrectinib (NTRK mutation) [90,91].

Allogeneic hematopoietic cell transplant (HCT) is rarely used and has not been well-studied for multiply relapsed LCH. A report of 87 patients (mostly children) with high-risk LCH who underwent HCT reported 73 percent OS for those transplanted since 2000 [92]. Estimated survival at three years was similar following myeloablative conditioning (MAC) and reduced intensity conditioning (RIC) (77 versus 71 percent, respectively), but relapses were more common after RIC (28 versus 8 percent).

Case reports and small case series suggest that HCT may be effective in preventing relapse among patients in complete remission at the time of HCT [92-97].

POST-TREATMENT SURVEILLANCE

Monitoring — Patients should be monitored periodically for relapse and treatment-related toxicity after completion of therapy.

We generally see patients every two to three months until a complete response is achieved and individualize the schedule of visits based on clinical judgment and comfort of the patient and clinician. Positron-emission tomography (PET)/computed tomography (CT) can be performed every two to three months until the best response, and the interval extended to six months or longer. However, the frequency and extent of follow-up visits should be individualized according to disease activity, treatment, and comfort of the clinician and patient.

The need for long-term follow-up for disease relapse was shown by a retrospective study of 335 patients with multisystem LCH who achieved a complete response; nearly half had disease reactivation within two years, but most again responded to therapy [72].

Patients with arginine vasopressin deficiency (AVP-D) and/or skull lesions in the orbit, mastoid, or temporal bone are at high risk of central nervous system (CNS) involvement at the time of initial diagnosis and at the time of relapse. These patients should have magnetic resonance imaging (MRI) without gadolinium contrast every one to two years for the 10 years after treatment [98]. Although radiologic changes may be present, treatment for progression in the CNS is usually reserved for those with worsening radiologic findings or evidence of clinical neurodegeneration.

Guidelines following treatment of childhood cancer or in those who have received chemotherapy have been published by the Children's Oncology Group, and are available at www.survivorshipguidelines.org [99].

Late effects — Long-term survivors can have residual abnormalities and late effects of the disease and its treatment.

The incidence of late effects is largely dependent on the extent of disease at diagnosis and the treatment received. Children with low-risk disease usually complete treatment with no long-term sequelae beyond mild obesity associated with prednisone therapy. Among 182 patients in a multi-institution study, 24 percent of patients with single-system LCH had permanent consequences, compared with 71 percent who presented with multisystem disease [100]. Among 71 children at a single institution, late sequalae were present in nearly two-thirds of patients who had >3 year follow-up, including dental problems, endocrine disorders, hearing loss, and CNS dysfunction [101]. It should be noted that patients in these two studies were often treated for only six months, which is shorter than the current standard of care.

It is presently unclear if the standard one year of continuation therapy will result in more or different late effects than previous treatments.

Examples include:

Growth and development – Growth and development problems are common (20 percent) in children who present at a young age; long-term prednisone therapy can be very toxic for very young children. Significant cognitive defects may develop in some long-term survivors [102], and hearing loss was found in 13 percent of survivors [100].

Endocrine abnormalities – Patients with AVP-D are at risk for panhypopituitarism and should be monitored carefully for adequate growth and development. (See "Clinical manifestations of hypopituitarism" and "Diagnostic testing for hypopituitarism".)

In a retrospective review of 141 patients with LCH and AVP-D, the 5- and 10-year risks of growth hormone (GH) deficiency were 35 and 54 percent, respectively [103]. There was no increased reactivation of LCH in patients who received growth hormone compared with those who did not.

A study of 144 patients with multisystem LCH who were followed in a late effects clinic reported that 35 percent had an endocrinopathy, including 49 of 50 with AVP-D [104]. Fifteen patients had received cranial irradiation for treatment of LCH causing AVP-D. GH deficiency occurred to 12 years (median 3.5 years) after diagnosis in 21 patients, 7 of whom had other anterior pituitary deficiencies; GH replacement was beneficial in some patients.

Infertility – Fertility preservation should be discussed, as there is a risk of infertility with cytotoxic chemotherapy. (See "Fertility and reproductive hormone preservation: Overview of care prior to gonadotoxic therapy or surgery".)

Second cancers – Patients with LCH can have second cancers before, coincident with, or after the diagnosis of LCH. It is uncertain if and how much treatment increases the risk for second cancers [105,106]. Acute myeloid leukemia and lymphoblastic lymphoma are reported [107,108]. Solid tumors reported to be associated with LCH include retinoblastoma, brain tumors, hepatocellular carcinoma, Askin tumor, and Ewing sarcoma.

Neurodegenerative LCH – Neurodegenerative LCH is a potentially catastrophic complication of brain involvement caused by infiltration of myeloid dendritic cells [55]. It can present with ataxia, behavioral dysfunction, and cognitive dysfunction more than a decade after initial diagnosis of LCH and is potentially irreversible. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis", section on 'Central nervous system'.)

Management of neurodegenerative LCH is discussed above. (See 'Non-BRAF V600E LCH' above.)

Other late effects – Neurologic problems, either secondary to vertebral compression or CNS-LCH (11 percent), and orthopedic defects from lesions of the femur, tibia, or humerus (20 percent) may be seen.

Liver disease may lead to ascending cholangitis, which is not amenable to any treatment other than liver transplant [109]. Patients with LCH-associated ascending cholangitis should be referred promptly for consideration of liver transplantation.

Chronic pain and fatigue – Patients with LCH often experience chronic pain and debilitating fatigue. The cause is uncertain and symptoms do not necessarily correlate with disease burden.

Many of these patients meet criteria for myalgic encephalomyelitis/chronic fatigue syndrome; these symptoms may be compounded by concomitant depression and anxiety. Acknowledging these symptoms is important and referral to appropriate specialties (eg, supportive care, fatigue clinic, psychiatry, psychology) may improve the quality of life. In some cases, use of oral stimulants like methylphenidate may be helpful and is considered safe for long-term use. (See "Clinical features and diagnosis of myalgic encephalomyelitis/chronic fatigue syndrome" and "Treatment of myalgic encephalomyelitis/chronic fatigue syndrome".)

CLINICAL TRIALS — An international directory of affiliated organizations dealing with the histiocytic disorders as well as the website for the Histiocytosis Association of America can be found at histio.org. The optimal therapy for all patients with LCH is to be enrolled on clinical trials. Information can be found at histiocytesociety.org or clinicaltrials.gov.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Histiocytic and dendritic cell neoplasms".)

SUMMARY AND RECOMMENDATIONS

Management – Management of Langerhans cell histiocytosis (LCH) is guided by (see 'Overview of management' above):

Single-system versus multisystem disease

Involvement of central nervous system (CNS) or a critical ("risk") organ (bone marrow, liver, or spleen)

Unifocal versus multifocal/extensive disease

Symptoms

Age – Preferred systemic treatment for children (≤20 years) (see 'Children' above) differs from adults. (See 'Adults' above.)

Single-system disease

Bone-only – Classification is described above. (See 'Bone' above.)

-Single bone – Curettage provides tissue diagnosis and treatment. Radiation therapy (RT) may be used for selected adults, but not children. (See 'Single bone lesion' above.)

-Multiple bones – For ≥2 bone lesions, lesion ≥5 cm, femoral or vertebral involvement, or CNS-risk bone (ie, orbit, mastoid, temporal, sphenoid), treatment involves systemic therapy. (See 'Systemic therapy' above.)

Surgery or RT may be added in selected cases. (See 'Adjunctive treatment' above.)

Skin-only – Topical steroids or mustard, or oral hydroxyurea, methotrexate, thalidomide, or lenalidomide can be effective. (See 'Skin' above.)

Primary pulmonary LCH is discussed separately. (See "Pulmonary Langerhans cell histiocytosis".)

Multisystem – Multisystem disease requires systemic therapy.

Children – For initial systemic treatment of children with LCH, we suggest induction therapy with vinblastine plus prednisone (V-P), rather than other chemotherapy regimens or a targeted agent (Grade 2C). (See 'Vinblastine-prednisone induction therapy' above.)

Treatment response guides further management; continuation therapy is 12 months for response to V-P. (See 'Continuation phase' above.)

Adults – Stratify treatment for adults according to involvement of CNS or risk organ (ie, liver, spleen, bone marrow):

-No CNS or risk organ involvement – For multisystem LCH in adults with no CNS or risk organ involvement, we suggest single-agent cytarabine or cladribine, rather than combination chemotherapy or a targeted agent (Grade 2C). (See 'No CNS or risk organ involvement' above.)

-CNS or risk organ involvement – For adults with BRAF V600E-mutated LCH and involvement of CNS or a risk organ, we suggest a BRAF inhibitor (eg, vemurafenib, dabrafenib), rather than systemic chemotherapy (Grade 2C). (See 'BRAF V600E LCH' above.)

For adults with BRAF wildtype LCH with CNS or risk organ involvement, we suggest cytarabine or cladribine, rather than combination chemotherapy or a targeted agent (Grade 2C). (See 'Non-BRAF V600E LCH' above.)

Response assessment – Positron emission tomography (PET) is preferred for response assessment, but computed tomography (CT), magnetic resonance imaging (MRI), or clinical assessment is used when PET is not available or appropriate (eg, brain lesions).

Relapsed disease – Treatment of relapsed/refractory LCH is individualized according to timing of relapse, sites of disease, performance status, and previous treatment. We generally treat with a different therapy from that used initially. (See 'Relapsed or refractory disease' above.)

Long-term surveillance – Patients are at risk for treatment-related toxicity, second cancers, and endocrine complications. (See 'Late effects' above.)

ACKNOWLEDGMENT — The UpToDate editorial staff acknowledges Laurence A Boxer, MD (deceased), who contributed to earlier versions of this topic review.

  1. 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 2014; 211:669.
  2. 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 2010; 184:4557.
  3. Luche H, Ardouin L, Teo P, et al. The earliest intrathymic precursors of CD8α(+) thymic dendritic cells correspond to myeloid-type double-negative 1c cells. Eur J Immunol 2011; 41:2165.
  4. Kim JR, Yoon HM, Jung AY, et al. Comparison of whole-body MRI, bone scan, and radiographic skeletal survey for lesion detection and risk stratification of Langerhans Cell Histiocytosis. Sci Rep 2019; 9:317.
  5. 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 2009; 52:97.
  6. Girschikofsky M, Arico M, Castillo D, et al. Management of adult patients with Langerhans cell histiocytosis: recommendations from an expert panel on behalf of Euro-Histio-Net. Orphanet J Rare Dis 2013; 8:72.
  7. Kim BE, Koh KN, Suh JK, et al. Clinical features and treatment outcomes of Langerhans cell histiocytosis: a nationwide survey from Korea histiocytosis working party. J Pediatr Hematol Oncol 2014; 36:125.
  8. Gadner H, Minkov M, Grois N, et al. Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis. Blood 2013; 121:5006.
  9. Howarth DM, Gilchrist GS, Mullan BP, et al. Langerhans cell histiocytosis: diagnosis, natural history, management, and outcome. Cancer 1999; 85:2278.
  10. Hamre M, Hedberg J, Buckley J, et al. Langerhans cell histiocytosis: an exploratory epidemiologic study of 177 cases. Med Pediatr Oncol 1997; 28:92.
  11. Jubran RF, Marachelian A, Dorey F, Malogolowkin M. Predictors of outcome in children with Langerhans cell histiocytosis. Pediatr Blood Cancer 2005; 45:37.
  12. Postini AM, Andreacchio A, Boffano M, et al. Langerhans cell histiocytosis of bone in children: a long-term retrospective study. J Pediatr Orthop B 2012; 21:457.
  13. Plasschaert F, Craig C, Bell R, et al. Eosinophilic granuloma. A different behaviour in children than in adults. J Bone Joint Surg Br 2002; 84:870.
  14. Laird J, Ma J, Chau K, et al. Outcome After Radiation Therapy for Langerhans Cell Histiocytosis Is Dependent on Site of Involvement. Int J Radiat Oncol Biol Phys 2018; 100:670.
  15. Atalar B, Miller RC, Dincbas FO, et al. Adult langerhans cell histiocytosis of bones : a rare cancer network study. Acta Orthop Belg 2010; 76:663.
  16. Arzoo K, Sadeghi S, Pullarkat V. Pamidronate for bone pain from osteolytic lesions in Langerhans'-cell histiocytosis. N Engl J Med 2001; 345:225.
  17. Farran RP, Zaretski E, Egeler RM. Treatment of Langerhans cell histiocytosis with pamidronate. J Pediatr Hematol Oncol 2001; 23:54.
  18. Sivendran S, Harvey H, Lipton A, Drabick J. Treatment of Langerhans cell histiocytosis bone lesions with zoledronic acid: a case series. Int J Hematol 2011; 93:782.
  19. Chellapandian D, Makras P, Kaltsas G, et al. Bisphosphonates in Langerhans Cell Histiocytosis: An International Retrospective Case Series. Mediterr J Hematol Infect Dis 2016; 8:e2016033.
  20. Morimoto A, Shioda Y, Imamura T, et al. Nationwide survey of bisphosphonate therapy for children with reactivated Langerhans cell histiocytosis in Japan. Pediatr Blood Cancer 2011; 56:110.
  21. Mammano S, Candiotto S, Balsano M. Cast and brace treatment of eosinophilic granuloma of the spine: long-term follow-up. J Pediatr Orthop 1997; 17:821.
  22. Kriz J, Eich HT, Bruns F, et al. Radiotherapy in langerhans cell histiocytosis - a rare indication in a rare disease. Radiat Oncol 2013; 8:233.
  23. Kotecha R, Venkatramani R, Jubran RF, et al. Clinical outcomes of radiation therapy in the management of Langerhans cell histiocytosis. Am J Clin Oncol 2014; 37:592.
  24. Ma J, Laird JH, Chau K, et al. Treatment Modality Choices and Outcome in Langerhans Cell Histiocytosis: Radiation Therapy Versus Other Modalities. Int J Radiat Oncol Biol Phys 2018; 102:E285.
  25. Krooks J, Minkov M, Weatherall AG. Langerhans cell histiocytosis in children: History, classification, pathobiology, clinical manifestations, and prognosis. J Am Acad Dermatol 2018; 78:1035.
  26. Minkov M, Prosch H, Steiner M, et al. Langerhans cell histiocytosis in neonates. Pediatr Blood Cancer 2005; 45:802.
  27. Lau L, Krafchik B, Trebo MM, Weitzman S. Cutaneous Langerhans cell histiocytosis in children under one year. Pediatr Blood Cancer 2006; 46:66.
  28. Simko SJ, Garmezy B, Abhyankar H, et al. Differentiating skin-limited and multisystem Langerhans cell histiocytosis. J Pediatr 2014; 165:990.
  29. Steen AE, Steen KH, Bauer R, Bieber T. Successful treatment of cutaneous Langerhans cell histiocytosis with low-dose methotrexate. Br J Dermatol 2001; 145:137.
  30. Womer RB, Anunciato KR, Chehrenama M. Oral methotrexate and alternate-day prednisone for low-risk Langerhans cell histiocytosis. Med Pediatr Oncol 1995; 25:70.
  31. Zinn DJ, Grimes AB, Lin H, et al. Hydroxyurea: a new old therapy for Langerhans cell histiocytosis. Blood 2016; 128:2462.
  32. Szturz P, Adam Z, Rehák Z, et al. Lenalidomide proved effective in multisystem Langerhans cell histiocytosis. Acta Oncol 2012; 51:412.
  33. McClain KL, Kozinetz CA. A phase II trial using thalidomide for Langerhans cell histiocytosis. Pediatr Blood Cancer 2007; 48:44.
  34. Hoeger PH, Nanduri VR, Harper JI, et al. Long term follow up of topical mustine treatment for cutaneous langerhans cell histiocytosis. Arch Dis Child 2000; 82:483.
  35. Lindahl LM, Fenger-Grøn M, Iversen L. Topical nitrogen mustard therapy in patients with Langerhans cell histiocytosis. Br J Dermatol 2012; 166:642.
  36. Ng SS, Koh MJ, Tay YK. Cutaneous Langerhans cell histiocytosis: study of Asian children shows good overall prognosis. Acta Paediatr 2013; 102:e514.
  37. Ravindran A, Goyal G, Failing JJ, et al. Florid dermatopathic lymphadenopathy-A morphological mimic of Langerhans cell histiocytosis. Clin Case Rep 2018; 6:1637.
  38. Allen CE, Merad M, McClain KL. Langerhans-Cell Histiocytosis. N Engl J Med 2018; 379:856.
  39. Gadner H, Grois N, Pötschger U, et al. Improved outcome in multisystem Langerhans cell histiocytosis is associated with therapy intensification. Blood 2008; 111:2556.
  40. Gadner H, Grois N, Arico M, et al. A randomized trial of treatment for multisystem Langerhans' cell histiocytosis. J Pediatr 2001; 138:728.
  41. 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 2018; 108:192.
  42. Cantu MA, Lupo PJ, Bilgi M, et al. Optimal therapy for adults with Langerhans cell histiocytosis bone lesions. PLoS One 2012; 7:e43257.
  43. Hazim AZ, Ruan GJ, Hu M, et al. Langerhans cell histiocytosis with lung involvement in isolation and multisystem disease: Staging, natural history, and comparative survival. Am J Hematol 2021; 96:1604.
  44. 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 2021; 96:E146.
  45. Dhall G, Finlay JL, Dunkel IJ, et al. Analysis of outcome for patients with mass lesions of the central nervous system due to Langerhans cell histiocytosis treated with 2-chlorodeoxyadenosine. Pediatr Blood Cancer 2008; 50:72.
  46. Barkaoui MA, Queheille E, Aladjidi N, et al. Long-term follow-up of children with risk organ-negative Langerhans cell histiocytosis after 2-chlorodeoxyadenosine treatment. Br J Haematol 2020; 191:825.
  47. 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 2009; 53:1271.
  48. Ng Wing Tin S, Martin-Duverneuil N, Idbaih A, et al. Efficacy of vinblastine in central nervous system Langerhans cell histiocytosis: a nationwide retrospective study. Orphanet J Rare Dis 2011; 6:83.
  49. Tazi A, Lorillon G, Haroche J, et al. Vinblastine chemotherapy in adult patients with langerhans cell histiocytosis: a multicenter retrospective study. Orphanet J Rare Dis 2017; 12:95.
  50. Duan MH, Han X, Li J, et al. Comparison of vindesine and prednisone and cyclophosphamide, etoposide, vindesine, and prednisone as first-line treatment for adult Langerhans cell histiocytosis: A single-center retrospective study. Leuk Res 2016; 42:43.
  51. Maia RC, de Rezende LM, Robaina M, et al. Langerhans cell histiocytosis: differences and similarities in long-term outcome of paediatric and adult patients at a single institutional centre. Hematology 2015; 20:83.
  52. Han X, Ouyang M, Duan M, et al. The combination of methotrexate and cytosine arabinoside in newly diagnosed adult Langerhans cell histiocytosis: a prospective phase II interventional clinical trial. BMC Cancer 2020; 20:433.
  53. Cao XX, Li J, Zhao AL, et al. Methotrexate and cytarabine for adult patients with newly diagnosed Langerhans cell histiocytosis: A single arm, single center, prospective phase 2 study. Am J Hematol 2020; 95:E235.
  54. Derenzini E, Fina MP, Stefoni V, et al. MACOP-B regimen in the treatment of adult Langerhans cell histiocytosis: experience on seven patients. Ann Oncol 2010; 21:1173.
  55. McClain KL, Picarsic J, Chakraborty R, et al. CNS Langerhans cell histiocytosis: Common hematopoietic origin for LCH-associated neurodegeneration and mass lesions. Cancer 2018; 124:2607.
  56. Haroche J, Cohen-Aubart F, Emile JF, et al. Dramatic efficacy of vemurafenib in both multisystemic and refractory Erdheim-Chester disease and Langerhans cell histiocytosis harboring the BRAF V600E mutation. Blood 2013; 121:1495.
  57. 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 2018; 4:384.
  58. Hazim AZ, Ruan GJ, Ravindran A, et al. Efficacy of BRAF-Inhibitor Therapy in BRAFV600E -Mutated Adult Langerhans Cell Histiocytosis. Oncologist 2020; 25:1001.
  59. Büchler T, Cervinek L, Belohlavek O, et al. Langerhans cell histiocytosis with central nervous system involvement: follow-up by FDG-PET during treatment with cladribine. Pediatr Blood Cancer 2005; 44:286.
  60. Kasper EM, Aguirre-Padilla DH, Alter RY, Anderson M. Histiocytosis X: Characteristics, behavior, and treatments as illustrated in a case series. Surg Neurol Int 2011; 2:57.
  61. Grois N, Pötschger U, Prosch H, et al. Risk factors for diabetes insipidus in langerhans cell histiocytosis. Pediatr Blood Cancer 2006; 46:228.
  62. Goyal G, Tazi A, Go RS, et al. International expert consensus recommendations for the diagnosis and treatment of Langerhans cell histiocytosis in adults. Blood 2022; 139:2601.
  63. 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 2010; 54:416.
  64. Durham BH, Lopez Rodrigo E, Picarsic J, et al. Activating mutations in CSF1R and additional receptor tyrosine kinases in histiocytic neoplasms. Nat Med 2019; 25:1839.
  65. Diamond EL, Durham BH, Ulaner GA, et al. Efficacy of MEK inhibition in patients with histiocytic neoplasms. Nature 2019; 567:521.
  66. Ballester LY, Cantu MD, Lim KPH, et al. The use of BRAF V600E mutation-specific immunohistochemistry in pediatric Langerhans cell histiocytosis. Hematol Oncol 2018; 36:307.
  67. Héritier S, Emile JF, Barkaoui MA, et al. BRAF Mutation Correlates With High-Risk Langerhans Cell Histiocytosis and Increased Resistance to First-Line Therapy. J Clin Oncol 2016; 34:3023.
  68. Awada G, Seremet T, Fostier K, et al. Long-term disease control of Langerhans cell histiocytosis using combined BRAF and MEK inhibition. Blood Adv 2018; 2:2156.
  69. Donadieu J, Larabi IA, Tardieu M, et al. Vemurafenib for Refractory Multisystem Langerhans Cell Histiocytosis in Children: An International Observational Study. J Clin Oncol 2019; 37:2857.
  70. Yang Y, Wang D, Cui L, et al. Effectiveness and Safety of Dabrafenib in the Treatment of 20 Chinese Children with BRAFV600E-Mutated Langerhans Cell Histiocytosis. Cancer Res Treat 2021; 53:261.
  71. Eckstein OS, Visser J, Rodriguez-Galindo C, et al. Clinical responses and persistent BRAF V600E+ blood cells in children with LCH treated with MAPK pathway inhibition. Blood 2019; 133:1691.
  72. Minkov M, Steiner M, Pötschger U, et al. Reactivations in multisystem Langerhans cell histiocytosis: data of the international LCH registry. J Pediatr 2008; 153:700.
  73. Imamura T, Sato T, Shiota Y, et al. Outcome of pediatric patients with Langerhans cell histiocytosis treated with 2 chlorodeoxyadenosine: a nationwide survey in Japan. Int J Hematol 2010; 91:646.
  74. Simko SJ, McClain KL, Allen CE. Up-front therapy for LCH: is it time to test an alternative to vinblastine/prednisone? Br J Haematol 2015; 169:299.
  75. Rosso DA, Amaral D, Latella A, et al. Reduced doses of cladribine and cytarabine regimen was effective and well tolerated in patients with refractory-risk multisystem Langerhans cell histiocytosis. Br J Haematol 2016; 172:287.
  76. Rodriguez-Galindo C, Jeng M, Khuu P, et al. Clofarabine in refractory Langerhans cell histiocytosis. Pediatr Blood Cancer 2008; 51:703.
  77. Abraham A, Alsultan A, Jeng M, et al. Clofarabine salvage therapy for refractory high-risk langerhans cell histiocytosis. Pediatr Blood Cancer 2013; 60:E19.
  78. 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 2014; 61:479.
  79. Bernard F, Thomas C, Bertrand Y, et al. Multi-centre pilot study of 2-chlorodeoxyadenosine and cytosine arabinoside combined chemotherapy in refractory Langerhans cell histiocytosis with haematological dysfunction. Eur J Cancer 2005; 41:2682.
  80. Apollonsky N, Lipton JM. Treatment of refractory Langerhans cell histiocytosis (LCH) with a combination of 2-chlorodeoxyadenosine and cytosine arabinoside. J Pediatr Hematol Oncol 2009; 31:53.
  81. 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 2015; 126:1415.
  82. Héritier S, Jehanne M, Leverger G, et al. Vemurafenib Use in an Infant for High-Risk Langerhans Cell Histiocytosis. JAMA Oncol 2015; 1:836.
  83. Héritier S, Hélias-Rodzewicz Z, Lapillonne H, et al. Circulating cell-free BRAFV600E as a biomarker in children with Langerhans cell histiocytosis. Br J Haematol 2017; 178:457.
  84. Váradi Z, Bánusz R, Csomor J, et al. Effective BRAF inhibitor vemurafenib therapy in a 2-year-old patient with sequentially diagnosed Langerhans cell histiocytosis and Erdheim-Chester disease. Onco Targets Ther 2017; 10:521.
  85. Kolenová A, Schwentner R, Jug G, et al. Targeted inhibition of the MAPK pathway: emerging salvage option for progressive life-threatening multisystem LCH. Blood Adv 2017; 1:352.
  86. Lee LH, Gasilina A, Roychoudhury J, et al. Real-time genomic profiling of histiocytoses identifies early-kinase domain BRAF alterations while improving treatment outcomes. JCI Insight 2017; 2:e89473.
  87. Montella L, Insabato L, Palmieri G. Imatinib mesylate for cerebral Langerhans'-cell histiocytosis. N Engl J Med 2004; 351:1034.
  88. Janku F, Amin HM, Yang D, et al. Response of histiocytoses to imatinib mesylate: fire to ashes. J Clin Oncol 2010; 28:e633.
  89. Wagner C, Mohme H, Krömer-Olbrisch T, et al. Langerhans cell histiocytosis: treatment failure with imatinib. Arch Dermatol 2009; 145:949.
  90. Taylor J, Pavlick D, Yoshimi A, et al. Oncogenic TRK fusions are amenable to inhibition in hematologic malignancies. J Clin Invest 2018; 128:3819.
  91. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020; 21:271.
  92. 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 2015; 169:711.
  93. Akkari V, Donadieu J, Piguet C, et al. Hematopoietic stem cell transplantation in patients with severe Langerhans cell histiocytosis and hematological dysfunction: experience of the French Langerhans Cell Study Group. Bone Marrow Transplant 2003; 31:1097.
  94. Kudo K, Ohga S, Morimoto A, et al. Improved outcome of refractory Langerhans cell histiocytosis in children with hematopoietic stem cell transplantation in Japan. Bone Marrow Transplant 2010; 45:901.
  95. Steiner M, Matthes-Martin S, Attarbaschi A, et al. Improved outcome of treatment-resistant high-risk Langerhans cell histiocytosis after allogeneic stem cell transplantation with reduced-intensity conditioning. Bone Marrow Transplant 2005; 36:215.
  96. Nagarajan R, Neglia J, Ramsay N, Baker KS. Successful treatment of refractory Langerhans cell histiocytosis with unrelated cord blood transplantation. J Pediatr Hematol Oncol 2001; 23:629.
  97. Suminoe A, Matsuzaki A, Hattori H, et al. Unrelated cord blood transplantation for an infant with chemotherapy-resistant progressive Langerhans cell histiocytosis. J Pediatr Hematol Oncol 2001; 23:633.
  98. Wnorowski M, Prosch H, Prayer D, et al. Pattern and course of neurodegeneration in Langerhans cell histiocytosis. J Pediatr 2008; 153:127.
  99. www.survivorshipguidelines.org (Accessed on May 19, 2011).
  100. Haupt R, Nanduri V, Calevo MG, et al. Permanent consequences in Langerhans cell histiocytosis patients: a pilot study from the Histiocyte Society-Late Effects Study Group. Pediatr Blood Cancer 2004; 42:438.
  101. Willis B, Ablin A, Weinberg V, et al. Disease course and late sequelae of Langerhans' cell histiocytosis: 25-year experience at the University of California, San Francisco. J Clin Oncol 1996; 14:2073.
  102. Nanduri VR, Lillywhite L, Chapman C, et al. Cognitive outcome of long-term survivors of multisystem langerhans cell histiocytosis: a single-institution, cross-sectional study. J Clin Oncol 2003; 21:2961.
  103. Donadieu J, Rolon MA, Pion I, et al. Incidence of growth hormone deficiency in pediatric-onset Langerhans cell histiocytosis: efficacy and safety of growth hormone treatment. J Clin Endocrinol Metab 2004; 89:604.
  104. Nanduri VR, Bareille P, Pritchard J, Stanhope R. Growth and endocrine disorders in multisystem Langerhans' cell histiocytosis. Clin Endocrinol (Oxf) 2000; 53:509.
  105. Egeler RM, Neglia JP, Puccetti DM, et al. Association of Langerhans cell histiocytosis with malignant neoplasms. Cancer 1993; 71:865.
  106. Egeler RM, Neglia JP, Aricò M, et al. The relation of Langerhans cell histiocytosis to acute leukemia, lymphomas, and other solid tumors. The LCH-Malignancy Study Group of the Histiocyte Society. Hematol Oncol Clin North Am 1998; 12:369.
  107. Goyal G, Shah MV, Hook CC, et al. Adult disseminated Langerhans cell histiocytosis: incidence, racial disparities and long-term outcomes. Br J Haematol 2018; 182:579.
  108. Ma J, Laird JH, Chau KW, et al. Langerhans cell histiocytosis in adults is associated with a high prevalence of hematologic and solid malignancies. Cancer Med 2019; 8:58.
  109. Braier J, Ciocca M, Latella A, et al. Cholestasis, sclerosing cholangitis, and liver transplantation in Langerhans cell Histiocytosis. Med Pediatr Oncol 2002; 38:178.
Topic 16627 Version 33.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟