LCH Sunil Sir

Embed Size (px)

Citation preview

  • 7/21/2019 LCH Sunil Sir

    1/25

    Langerhans cell histiocytosis: Current insights in a molecular age with

    emphasis on clinical oral and maxillofacial pathology practice

    John Hicks, DDS, MS, PhD, MD,a and Catherine M. Flaitz, DDS, MS,b Houston, Tex

    TEXAS CHILDRENS HOSPITAL, BAYLOR COLLEGE OF MEDICINE, AND THE UNIVERSITY OF TEXASHEALTH SCIENCE CENTER AT HOUSTON

    Langerhans cell histiocytosis (LCH) commonly involves the oral and maxillofacial region, and comes to the attention

    of dental practitioners when a patient presents with orofacial pain and a bony or soft tissue lesion. This is a relatively rare entity,

    which has made it difficult to investigate the clinical, biologic, and molecular aspects of the disease. Treatment protocols are not

    well defined, particularly in adults. During the past decade, the Histiocyte Society has formulated various LCH categories, based

    on risk stratification, and treatment protocols for the pediatric population. Adult trials are currently available through the

    Histiocyte Society. Although there has been considerable controversy, the neoplastic nature of LCH has been established by

    demonstrating clonality. LCH symptoms and the development and persistence of LCH lesions have been ascribed to a

    chemokine/cytokine storm due to autocrine and paracrine mechanisms. Discovery of biologic, cytogenetic, and molecular

    abnormalities in LCH have already affected treatment by providing novel therapeutic targets. (Oral Surg Oral Med Oral Pathol

    Oral Radiol Endod 2005;100:S42-66)

    Langerhans cell histiocytosis is rare and unique

    among human diseases. Our understanding of this

    condition has significantly advanced and evolved over

    the past century.1,2 Recognition of the disease entity

    took a considerable period of time. Although not

    recognized as a histiocytic disease, the first case was

    described in 1865 in a 4-year-old child with impetigo

    andlarge punched-out osteolytic lesions in his calvar-

    ium.1,2 At that time, the bony lesions were considered to

    be congenital in nature, and the child died 1 month later

    of respiratory compromise. The clinical entity Hand-Schuller-Christian disease came about with independent

    case reports of children with exophthalmos, polyuria,

    great thirst (polydypsia), osteomalacia, and map-like

    skull defects by Hand in 1892, Schuller in 1915, and

    Christian in 1919.1,2 The disease was initially attributed

    to tuberculosis. Letterer-Siwe Disease was initially

    described in 1924 by Letterer and later in 1933 by Siwe

    as an acute fulminant nonleukemic disorder of the

    reticuloendothelial system in 2 young children.1,2 This

    condition was characterized by marked splenomeg-

    aly, hepatomegaly, lymphadenopathy, localized bone

    tumors, hemorrhagic diathesis, anemia, and hyperplasia

    of nonlipidized histiocytes. This disease was consid-

    ered to be a rare unknown storage disorder. Eosinophilic

    granuloma of bone was described first in 1930 by

    Mignon as a granulomatous bone lesion in an adolescent

    boy.1,2 It was Lichenstein and Jaffe in 1940 who coined

    the term eosinophilic granuloma of bone, and these

    lesions were thought to represent viral granulomasdespite their propensity to destroy bone.1,2

    The suggestion that Hand-Schuller-Christian disease,

    Letterer-Siwe disease, and eosinophilic granuloma of

    bone were part of thesame disease process with variation

    in severity, site of involvement, and stage was made

    by Farber in 1941.1,2 Lichenstein and Jaffe embraced

    this idea, and in 1953 included allthese entities under the

    general category of Histiocytosis X.1-3 This term was

    intended to reflect the inflammatory and proliferativenature of the disease. The unification of these three

    separately described diseases under one category was

    based on the nearly identical histopathologic featuresof the lesions, which were composed of eosinophils,

    histiocytes, and lymphocytes. Histiocytosis X was sub-

    divided into acute and subacute disseminated, chronic

    disseminated, and localized forms. The original disease

    designations, Hand-Schuller-Christian disease, Letterer-

    Siwe disease, and eosinophilic granuloma, are still used

    by some practitioners when referring to the clinical

    features of Langerhans cell histiocytosis (LCH).

    In the inaugural 1948 volume ofOral Surgery, Oral

    Medicine, and Oral Pathology,which has since evolved

    aProfessor of Pathology, Medical Director of Surgical and Ultra-

    structural Pathology, Texas Childrens Hospital; Medical Director ofTexas Childrens Cancer Center, Cytogenetics and Molecular

    Genetics, Baylor College of Medicine; and Adjunct Professor,

    Department of Pediatric Dentistry, The University of Texas Health

    Science Center at Houston, Dental Branch, Houston, Tex.bDean and Professor, Departments of Diagnostic Sciences and

    Pediatric Dentistry, The University of Texas Health Science Center

    at Houston, Dental Branch, Houston, Tex.

    Received for publication Jun 14, 2005; returned for revision Jun 19,

    2005; accepted for publication Jun 24, 2005.

    1079-2104/$ - see front matter

    2005 Mosby, Inc. All rights reserved.

    doi:10.1016/j.tripleo.2005.06.016

    S42

    http://-/?-http://-/?-http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    2/25

    into the current journal, eosinophilic granuloma of the

    jaw in a 31-year-old man with an anal fistula was

    chronicled.3 From 1942 until 1945, the patient under-

    went several oral surgical procedures with biopsies

    that revealed features that would now be recognized as

    LCH, but were referred to as an unusual inflammatory,

    infectious, and peculiar granulomatous process. Similarfindings were noted in the anal ulcer. It was not until late

    1945 that the quite probable diagnosis of eosinophilic

    granuloma of bone was rendered. Radiation therapy was

    initiated and the jaw lesions resolved with no evidence

    of disease at a 1-year follow-up.

    Although LCH is a rare orphan disease, consider-

    able advances have been made since the first case report

    of eosinophilic granuloma of the jaw occurred in the

    1948 volume ofOral Surgery, Oral Medicine, and Oral

    Pathology.3 Clinical classification, advances in clinical

    and pathologic diagnostic methods, identification of the

    responsible cell type, and characterization of cellular

    components and molecular genetics in this disease

    process have been further defined.

    CELL OF ORIGIN: LANGERHANS CELLHISTIOCYTES

    Almost 140 years ago, Langerhans cells in the skin

    were discovered using gold labeling techniques and

    described as being intraepidermal receptors for cutane-

    ous nervous system signals.1,2 Little progress was made

    in understanding the nature of Langerhans cells until the

    early 1960s when electron microscopy became avail-

    able. Definitive identification of Langerhans cells was

    possible with characterization of a cell-specific organ-elle, the Langerhans body, and the histiocytic nature of

    the cell became apparent.1,2 This unique ultrastructural

    organelle is commonly known as the Birbeck granule.

    With the development of immunocytochemical anti-

    body techniques, in situ hybridization, and molecular

    techniques, a great deal of knowledge regarding these

    cells has been gained. Langerhans cells have proven to

    be bone marrowederived antigen processing cells and

    represent the most peripheral extension of the immune

    system. These cells were considered to be histiocytes.

    The pathologic proliferation of these cells is referred to

    as LCH.Langerhans cells are derived from CD34-positive

    (1) hematopoietic myeloid stem cells in the bone

    marrow.4-15 Under the influence of various immune

    modulators, the progenitor cells can be induced to

    become typical macrophages (histiocytes, monocytes)

    or transitional macrophages/dendritic cell precursors.

    These precursor cells give rise to a variety of dendritic

    cells when specific inducing cytokines, chemokines,

    and growth factors are present. Langerhans cells, in-

    determinant cells, interdigitating dendritic cells, dermal

    dendrocytes, and follicular dendritic cells arise from

    these transitional macrophage/dendritic precursors.

    Mesenchymal progenitor cells may further influence

    differentiation toward dermal dendrocytes and lymph

    node follicular dendritic cells.

    With the advent of immunocytochemical and flow

    cytometric methods, it has been possible to defineseveral different disease entitiesbased on the cell types

    and clinical features (Table I).4-15 The contemporary

    classification of histiocytic disorders is based on the

    cell of origin responsible for the disease, relatively well-

    characterized clinical features, the biologic behavior

    of the disease, and proven malignant potential. LCH

    is classified under dendritic cellerelated disorders of

    varied biologic behavior, and the responsible cell is con-

    sidered to be an immature dendritic cellthe Langer-

    hans cell histiocyte. It is interesting to note that had the

    dendritic origin of the proliferating cells in LCH been

    recognized during Lichenstein and Jaffes era, this

    disease would probably be known currently as Langer-

    hans cell dendritocytosis.

    LANGERHANS CELLS AND LANGERHANS CELLHISTIOCYTES: A COMPARISON

    Langerhans cells are freely mobile cells that originate

    from bone marrow myeloid precursors, and migrate via

    afferent lymphatic channels to populate the epidermis,

    regional lymph nodes, thymic epithelium, and oral and

    bronchial mucosa.1,2,4-15 Although these cells represent

    less than 2% of the epidermal cell population, their

    dendritic processes cover 25% of all epithelial cell

    surfaces in the epidermis. Each Langerhans cell pos-sesses many dendritic processes that may overlap with

    processes from other Langerhans cells. The Langerhans

    cell is a mature dendritic cell that has important immu-

    nologic functions. These cells are antigen-presenting

    cells, and are capable of presenting alloantigens and

    soluble foreign antigens to naive helper T-cell lympho-

    cytes (CD4 positive). Langerhans cells are nonadherent

    and do not proliferate in normal tissue culture condi-

    tions. Even though these cells can endocytose soluble

    antigens, they are considered to be nonphagocytic cells.

    With the uptake of antigen, Langerhans cells migrate

    to afferent lymphatic channels and make their way toparacortical zones in regional lymph nodes. Once there,

    the endocytosed antigens are presented to naive helper

    T cells. Keratinocytes produce certain cytokines that

    can inhibit normal Langerhans cell function. Ultraviolet

    light reduces their antigen-presenting function and may

    be a primary factor in skin cancer development. Langer-

    hans cells may be induced to express CD4 on their

    surfaces. This allows for infection by human immuno-

    deficiency virus (HIV), andLangerhans cells are consid-

    ered a reservoir for this virus. Strong chemoattractants

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S43

  • 7/21/2019 LCH Sunil Sir

    3/25

    that induce migration of Langerhans cells include

    interleukin (IL)-1-beta, IL-8, and granulocyte macro-

    phage-colony stimulating factor (GM-CSF). Langer-

    hans cells also produce IL-1-beta and tumor necrosis

    factor (TNF)-alpha. Langerhans cells have not proven to

    be clonal. These cells are primary antigen processing

    cells, and represent the most peripheral extension of the

    immune system.

    In contrast to normal Langerhans cells, Langerhans

    cell histiocytes are immature dendritic cells that areresponsible for a rare, unique disease process

    Langerhans cell histiocytosis.1,2,4-15 These histiocytes

    are a constituent of lesions occurring in bone, skin,

    lymph nodes, spleen, liver, thymus, bone marrow, the

    central nervous system, and the gastrointestinal tract.

    Despite the activated state of Langerhans cell histio-

    cytes, their ability to present antigens is either absent

    or rarely observed. These cells do not have the ability

    to migrate from the lesional tissue, and along with

    inflammatory cells recruit other cells to thelesion. These

    unique histiocytes lack dendritic cell processes and

    have a round to ovoid, epithelioid morphology. Typical

    cytokines produced by Langerhans cell histiocytes

    include IL-1-beta, IL-3, IL-4, IL-8, TNF-alpha, and

    GM-CSF. These cytokines provide an autocrine, as well

    as a paracrine, function in lesion establishment and

    persistence. These cells have proven to be clonal.Langerhans cell histiocytes are immature dendritic cells

    that lack the ability to be functional antigen-presenting

    cells.

    The common origin of Langerhanscells andLangerhans

    cell histiocytes1,2,4-15 is based on the discovery of Birbeck

    granules with transmission electron microscopy. Only

    these 2 cell types possess such pentalaminar structures.

    Extensive immunocytochemical and flow cytometric

    analyses have also provided additional support for the

    shared origin of these 2 cell types. Both express S100

    protein, HLA-DR, CD1a, and CD207 (langerin), while

    being negative for macrophage markers (CD68, lyso-

    zyme, CD14, CD163).

    LANGERHANS CELL HISTIOCYTOSIS:GENERAL OVERVIEWClassification schemes

    Several clinical classifications of LCH are used

    by many practitioners (Table II and Table III).4-24

    Eosinophilic granuloma represents the most common

    form of this disease and is typically a localized, unifocal

    osteolytic lesion. Older children and adults are com-

    monly affected, but most patients are younger than 20

    years of age. Multifocal lesions are less common and

    skin lesions are quite rare. Letterer-Siwe disease affectsinfants, with mucocutaneous lesions being common.

    Seborrheic dermatitiselike lesions, ulcers, and purpuric

    nodules of the skin and subcutis occur at multiple sites.

    The lung, liver, and spleen may be involved also. Hand-

    Schuller-Christian disease is characterized by a classic

    triad with osteolytic lesions, exophthalmos, and diabe-

    tes insipidus. Those affected tend be young children.

    A unique form of LCH is congenital and typically

    undergoes involution over time. Congenital self-healing

    LCH presents in the neonate or young infant as viola-

    ceous red-brown firm nodules (Table II).18,21,25 Such a

    pattern of clinical presentations may raise concernregarding the possibility of congenital leukemia and

    neuroblastoma or an infectious process. The presenta-

    tion at birth or shortly after is beneficial in identifying

    this rare entity and avoiding unnecessary medical man-

    agement. Certain ultrastructural features are specific to

    the congenital self-healing form of LCH. Young adult

    smokers have a unique form of LCH with pulmonary

    involvement only (Table II andTable III).4,17,22-24 No

    other lesional sites are found in these individuals. The

    major sequelae of pulmonary LCH are progressive

    Table I. Contemporary classification of histiocyticdisorders*

    Disorders of varied biological behavior

    Dendritic cellrelated disorders

    Langerhans cell histiocytosis

    Secondary dendritic processes (association with Hodgkin

    lymphoma, acute lymphoblastic leukemia, acute

    myelogenous leukemia)Juvenile xanthogranuloma and related disorders

    Solitary histiocytomas of various dendritic cell phenotypes

    Macrophage-related disorders

    Hemophagocytic syndromes

    Primary hemophagocytic lymphohistiocytosis

    (familial, sporadic, and viral infection associated)

    Secondary hemophagocytic lymphohistiocytosis

    Infection associated

    Malignancy associated

    Other

    Rosai-Dorfman disease

    (sinus histiocytosis with massive lymphadenopathy)

    Solitary histiocytoma with macrophage phenotype

    Malignant disordersMonocyte-related disorders

    Leukemias

    Acute monocytic leukemia (FAB M5A and M5B)

    Acute myelomonocytic leukemia (FAB M4)

    Chronic myelomonocytic leukemia

    Extramedullary monocytic tumor of sarcoma

    (monocyte-derived granulocytic sarcoma)

    Dendritic cellrelated histiocytic sarcoma

    Follicular dendritic cell sarcoma

    Interdigitating dendritic cell sarcoma

    Dendritic cell sarcomas (based on phenotype)

    Macrophage-related histiocytic sarcoma

    *Modified from reference4: Working Group of The Histiocyte Society

    and World Health Organization Committee on Histiocytic/ReticulumCell Proliferations.

    OOOOE

    S44 Hicks and Flaitz August 2005

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    4/25

    interstitial fibrosis and respiratory compromise. There is

    an association with malignant tumor development. This

    disease is extremely rare in children.

    The most current method for the categorization of

    LCH is based on risk stratification of affected individ-

    uals into recommended treatment protocols formulatedby the Histiocyte Society.4,17 The Histiocyte Society has

    placed LCH into several disease groups (Table II).

    Unifocal disease involves a single disease system with a

    single site of involvement. There is a good prognosis in

    this group, typically composed of older children and

    adults. Multifocal single system disease is present when

    several lesions are identified in a single organ system.

    Bone is most commonly involved, and this representsmultifocal eosinophilic granuloma. The prognosis is

    intermediate in this group that typically involves young

    children. The worst prognosis is found in the multifocal

    multisystem disease group. Multiple lesions are found inmore than one organ group. Commonly affected organ

    systems include bone, skin, liver, spleen, and lymph

    nodes. Children younger than 2 years of age represent

    the majority of patients in this group. The Histiocyte

    Society also recognizes congenital self-healing and

    pulmonary-only LCH groups. The final disease group is

    secondary LCH that is associated with a variety of

    tumors. The malignancies most frequently associated

    with LCH are leukemias and lymphomas, and less

    commonly sarcomas and carcinomas.4,17,26

    Table II. Langerhans cell histiocytosis: clinical typesand categorization schemes*

    Clinical types of Langerhans cell histiocytosis

    Eosinophilic granuloma

    Most common form of Langerhans cell histiocytosis

    Localized form, most benign

    Older children and adults

    [75% of affected individuals younger than 20 years of ageUnifocal lesions 3 times more common

    (skull[ femur[pelvis[vertebra[jaws)

    Multifocal lesions less common (50% skull, 16% jaws)

    Rare skin lesions

    Letterer-Siwe disease

    Usually 1st year of life

    Mucocutaneous lesions including gingiva and oral mucosa

    Seborrheic dermatitis-like skin lesions

    Purpuric red-brown nodules

    Ulcerated painful nodules involving perineal, inguinal,

    retroauricular, and external auditory canal regions

    Lung, liver, and spleen involvement

    Hand-Schuller-Christian disease

    Usually 2- to 6-year-old children

    Classic triad: osteolytic lesions, exophthalmos, and

    diabetes insipidus

    Skin and oral lesions

    Congenital self-healing Langerhans cell histiocytosis

    (reticulohistiocytosis, Hashimoto-Pritzker disease)

    Pulmonary Langerhans cell histiocytosis

    Categorization by Histiocyte Society for treatment

    protocols (current)

    Unifocal disease

    Single system disease with single site of involvement

    Most commonly bone

    Older children and adults

    Good prognosis

    Multifocal single system disease

    Multiple sites of involvement in single organ system

    Most commonly bone

    Young children

    Intermediate prognosis

    Multifocal multisystem disease

    Multiple involved sites in more than one organ system

    Most commonly bone, skin, liver, spleen, and lymph nodes

    Children younger than 2 years of age and infants

    Poor prognosis

    Congenital self-healing Langerhans cell histiocytosis

    Multiple skin lesions at birth or shortly after mimicking

    congenital neuroblastoma or leukemia (blueberry

    muffin baby)

    Neonates and infants

    Self-healing involutionPulmonary Langerhans cell histiocytosis

    Young adult smokers (smokers malady)

    Indolent progression to pulmonary fibrosis

    Strong association with malignancies

    Extremely rare in children

    Secondary Langerhans cell histiocytosis associated

    with neoplasms

    Acute lymphoblastic and myelogenous leukemias

    Chronic myelogenous leukemia

    Myelodysplastic disorder association

    Non-Hodgkin and Hodgkin lymphoma

    Retinoblastoma

    Table II. Continued

    Osteosarcoma

    Thyroid carcinoma

    Lung cancer (adenocarcinoma, small cell carcinoma)

    Prostate cancer

    Breast cancer

    Parathyroid adenoma

    Pancreatic cystadenoma

    Categorization by extent of disease

    Restricted Langerhans cell histiocytosis

    1. Skin rash with no other sites of involvement

    (biopsy proven)

    2. Monostotic bone lesions with or without diabetes

    insipidus, regional lymph node involvement, or skin rash

    3. Polyostotic bone lesions, consisting of several different

    bones or \2 bone lesions in a single bone, with or without

    diabetes insipidus, regional lymph node involvement, or skin

    rash

    Extensive Langerhans Cell Histiocytosis

    1. Visceral organ involvement with or without bone

    involvement, diabetes insipidus, regional lymph node

    involvement or skin rash, and without signs of organdysfunction

    2. Visceral organ involvement with or without bone

    involvement, diabetes insipidus, regional lymph node

    involvement or skin rash, and with signs of organ

    involvement of lung, liver or hematopoietic system

    *Compiled from references4 to 24.

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S45

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    5/25

    LCH has also been categorized according to extent of

    disease (Table II).2,20 Restricted and extensive cate-

    gories have been defined. The restricted LCH category is

    composed of skin rash only, monostotic bone lesion, and

    polyostotic bone lesion subcategories, as detailed in

    Table II. The extensive LCH category is based on vis-ceral organ involvement with or without organ dysfunc-

    tion. As expected, outcome is worse in those with

    extensive disease versus those with restricted diseases.

    Treatment decisions may be based on this categorization

    system with some protocols.

    EpidemiologyLCH is a rare disease that affects 5 children per

    million population and about 1 to 2 adults per million

    population.1,2,4-6,9,15-17,22-25 It is predominantly a child-

    hood disease with more than 50% of affected individuals

    being younger than 15 years. The peak incidence is

    between 1 and 4 years of age. This disease (Table III)

    affects a young agepopulation (mean age27 years), with

    a young adult smoker population affected by pulmonary-

    only disease (mean age 41 years) and primarily neonates

    Table III. Langerhans cell histiocytosis: clinical fea-tures*

    Distribution by category, %

    Unifocal disease 36

    Multifocal single system disease 33

    Multifocal multisystem disease 31

    Age (mean)

    Langerhans cell histiocytosis (all) 27 yUnifocal single system disease 18 y

    Multifocal single system disease 26 y

    Multifocal multisystem disease 25 y

    PulmonaryLangerhanscellhistiocytosis 41 y

    Congenital self-healing Langerhans

    cell histiocytosis

    5.2 d

    Langerhans cell histiocytosis (all)

    Age range Neonate to 83 y

    \20 years of age 42%

    Gender, M:F

    Langerhans cell histiocytosis (all) 1.0:1.1

    Pulmonary Langerhanscell histiocytosis 1.0:2.2

    Congenital self-healing Langerhans

    cell histiocytosis

    1.0:1.0

    Presenting clinical features, %

    Local bone pain 41

    Dyspnea 14

    Malaise 9

    Abnormal chest x-ray 9

    Painful scalp mass 7

    Pneumothorax 6

    Diabetes insipidus 5

    Scalp rash 3

    Skin rash on trunk 3

    Head and neck lymphadenopathy 2

    Otitis media 2

    Mucous membrane ulcer 2

    Orbital proptosis 2

    Chronic cough 1Scrotal mass \1

    Pathologic fracture \1

    Loose teeth \1

    Cor pulmonale \1

    Bony involvement sites, %

    Head and neck

    Skull 27-43

    Mandible 7-9

    Maxilla 1

    Cervical vertebra 2

    Extremities

    Lower extremity, proximal 14-15

    Upper extremity, proximal 6-7

    Lower extremity, distal 2-3

    Upper extremity, distal 1-2Ribs 10-14

    Pelvis 9-12

    Thoracic, lumbar, and sacral

    vertebrae

    5-10

    Scapula 5

    Clavicle 3

    Treatment and outcome

    Unifocal bone disease Local Excision with or

    without radiation

    11% Relapse

    97% Disease-freesurvival

    Table III. Continued

    Multifocal bone disease only Local excision with or

    without radiation

    and/or chemotherapy

    76% Relapse

    91% Disease-freesurvival

    6% Alive with disease

    3% Died of diseaseMultifocal multisystem disease Combination surgery, che-

    motherapy, and radiation

    95% Relapse

    74% Disease-freesurvival

    11% Alive with disease

    15% Died of disease

    Pulmonary disease only Prednisone and/or

    chemotherapy and/or

    surgery

    85% Disease-freesurvival

    4% Alive with

    progressive disease

    11% Died of disease

    Morbidity: lifelong sequelae

    Diabetes iInsipidus

    Growth hormone deficiency

    Orthopedic problems

    Pulmonary fibrosis

    Biliary cirrhosis

    Cerebellar ataxia

    Cognitive dysfunction

    Mortality with or without risk

    organ involvement (lung, liver,

    bone marrow, spleen), %

    Without risk organ involvement 10

    With risk organ involvement 30-50

    *Compiled from references7, 16, and 17.

    OOOOE

    S46 Hicks and Flaitz August 2005

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    6/25

    with congenital self-healing disease (mean age 5.2

    days). Unifocal single system, multifocal single system,

    and multifocal multisystem disease tend to be nearly

    equally represented (Table III). The gender ratio is equal

    in all LCH categories, with the exception of pulmonary-

    only disease, where women are affected more than twice

    as often.The most commonpresenting symptom is local bone

    pain (41%,Table III).7,16,17 Various symptoms based on

    organ system involvement have been noted (Table III).

    Of interest for the head and neck region are painful scalp

    mass (6%), scalp rash (3%), cervical lymphadenopathy

    (2%), otitis media (2%), mucous membrane ulcer (2%),

    orbital proptosis (2%), and, rarely, loose teeth (\1%).

    Despite the low proportion of symptoms in the head and

    neck region, bony involvement by LCH is quite com-

    mon (Table III). The skull and mandible are frequently

    affected, as well.

    Treatment of LCH has been quite variable during

    the past 50 years, and there has been a lack of coordi-

    nated treatment protocols, particularly for adults (Table

    III).2,7,16 With unifocal bone disease, local excision withor without radiation therapy has led to more than 95%

    disease-free survival. However, relapse occurs in about

    10% of cases. Multifocal bone disease may have similar

    surgical treatment with or without the addition of

    chemotherapy. Relapses are quite frequent with multi-

    focal bone disease (76%). However, disease-free sur-

    vival can be achieved in most of the affected patients

    (91%). A small proportion of individuals die of their

    disease (3%). Treatment of multifocal multisystem

    disease employs the combination of surgery, chemo-therapy, and radiation therapy. The vast majority of

    individuals have relapses (95%). One sixth will die of

    disease. Disease-free survival is achieved by about 75%

    of affected individuals. With pulmonary disease only,

    treatment is quite variable ranging from prednisone

    therapy to chemotherapy to surgery for lesions com-

    promising the respiratory tract. A certain percentage of

    individuals die of disease (11%). Disease-free survival

    is noted in 85%. This form of LCH may have ongoing

    progressive interstitial fibrosis in many affected pa-

    tients, even when tobacco use is discontinued.

    LCH produces lifelong effects, the nature of whicharedependent on the organ system(s) involved (Table

    III).7,16,17 In particular when there is hypothalamic/pituitary gland involvement, the affected individual

    may experience diabetes insipidus and growth hormone

    deficiency. Central nervous system lesions may lead to

    cerebellar ataxias with gait and mobility abnormalities

    and cognitive dysfunction. With extensive or multifocal

    bony lesions, skeletal growth and orthopedic abnor-

    malities may be seen. The liver is particularly suscep-

    tible to biliary cirrhosis due to bile duct damage and

    sclerosis caused by the infiltration of Langerhans cell

    histiocytes.

    Certain organ systeminvolvement increases the risk

    for mortality (Table III).7,16,17 High-risk organs include

    the lung, liver, bone marrow, and spleen. Death occurs in

    30% to 50% of those with high-risk organ involvement,

    comparedwith only 10% without lesions in these organs.Certain factors are predictive of progressive

    disease in LCH (Table IV).7,16,17 Several factors have

    an extremely high relative risk for progressive disease,

    such as osseous and mucocutaneous disease, osseous

    and extraosseous disease, and treatment relapse with

    multifocal multisystem disease. The presence of these

    factors necessitates aggressive therapy. In the pediatric

    age group, risk stratification for more intensive treat-

    ment is based on sites of involvement.17 Low-risk sites

    are defined as skin, bone, lymph nodes, and pituitary

    glands. High-risk sites include lung, liver, bone marrow,

    and spleen. Special sites that demand more aggressivetherapy are the mastoid bone, orbit, temporal bone, and

    central nervous system.

    Children and adults show different patterns of

    involvement by LCH (Table V).16,17,22-24 Bone disease

    is extremely frequent in children and less frequent in

    adults. The jaws are involved in 30% of adults, but in

    less than 10% of children. Skull lesions are about twice

    as common in children. Skin disease is seen with equal

    frequency. Pulmonary disease either in multifocal or

    isolated LCH is very frequent in adults and infrequent

    Table IV. Predictive factors in progression of Lang-erhans cell histiocytosis and low-risk and high-riskcategories for pediatric Langerhans cell histiocytosis*

    Predictive factors in progression Relative risk

    Osseous and mucocutaneous disease 40.7

    Osseous and extraosseous disease 37.3

    Treatment relapse with multifocal

    multisystem disease

    37.2

    Osseous disease involving 3 or more bones 6.1

    Mucous membrane disease 5.1

    Hepatosplenomegaly in patient younger

    than 3 years old

    4.5

    Pituitary-hypothalamic axis involvement

    and multisystem disease

    2.2

    Younger than 5 years of age at presentation 2.1

    3 or more organ systems involved by disease 1.8

    Pediatric categorization for therapy

    Low-risk sites: skin, bone, lymph nodes,

    and pituitary

    High-risk sites: lung, liver, bone marrow,

    and spleen

    Special site lesions: skull (mastoid, orbit,

    temporal bone; associated with diabetes

    insipidus and parenchymal brain lesions

    with neurodegenerative process)

    *Compiled from references16, 17.

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S47

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    7/25

    to rare in children. Genital lesions are frequent in adults

    and rare in children. Diabetes insipidus is found fre-

    quently in both children and adults.

    ORAL AND MAXILLOFACIAL LANGERHANSCELL HISTIOCYTOSIS

    LCH involves the head and neck region quite

    commonly, and, in particular, the bones of the skull

    and jaws (Table IV, Table V, Figs 1-4).27-35 With oral and

    maxillofacial LCH (Table V, Fig 1-4), more than 90% of

    affected individuals are younger than 40 years of age,

    with a mean age of about 19 years. Unifocal single

    system disease represents about 50% of maxillofacial

    LCH, with multifocal single system disease and mul-

    tifocal multisystem disease equally distributed in the

    remaining 50% of cases. The jaws are involved twice

    as frequently as the oral soft tissues. The mandible is

    3 times more frequently affected than the maxilla. Theposterior regions of the jaws are more frequently in-

    volved than the anterior regions. Oral soft tissue lesions

    are most commonly found with the gingiva and hard

    palate. The floor of the mouth, maxillary sinus, and

    buccal mucosaeach account forless than 10% of lesions.

    The most common oral cavity signs and symptoms at

    presentation are intraoral mass, pain, gingivitis, loose

    teeth, oral mucosal ulcer, impaired healing, and halito-

    sis. Extraoral signs and symptoms are quite common,

    and include soft tissue and bone lesions outside the oral

    cavity. Diabetes insipidus, lymphadenopathy, anemia,

    skin lesions, exophthalmos, and hepatomegaly are lesscommon. The skull and lower extremities are the most

    common extraoral sites of bony involvement.

    Jaw lesions tend to have a unilocular radiolucent

    appearance (Table VI, Fig 3), with well-demarcated

    borders in two thirds of the cases and poorly defined

    borders in the remainder.27-35 Tooth displacement is

    associated with about half of the bony lesions. Root

    resorption is seen less frequently.

    Surgical management alone is used in 50% of cases

    with an additional 23% of the lesions being treated with

    both surgery and radiation therapy.27-35 Radiation alone

    is used in one fifth of affected individuals. Chemo-

    therapy and intralesional steroid injection are infre-

    quently employed.

    Recurrence of LCH is variable, but similaramong the

    various disease categories (Table VI). 27-35 A single

    recurrence is noted in 15% and a second recurrence inless than 5% of patients. Treatment modalities vary in

    the proportion of cases with recurrences. Chemotherapy

    has had no reported recurrences, however this form of

    therapy has not been used frequently enough to assess its

    effect accurately. Surgery alone has a 12% recurrence

    rate, compared with 25% for radiation therapy alone and

    19% for combined surgery and radiation therapy.

    Survival in oral and maxillofacial LCH is quite

    favorable with only 7% of patients dead from disease

    (Table VI).27-35 Following treatment, there was no

    evidence of disease in slightly over half of the patients;

    while 17% were alive with disease.

    PATHOLOGIC FEATURES OF LANGERHANSCELL HISTIOCYTOSISLight microscopic features

    The histopathologic features of LCH are well char-

    acterized and recognized readily by oral and maxillo-

    facial pathologists (Fig 5).1,2,4-7,9-12,21,36-38 The typical

    lesion is composed of an admixture of Langerhans cell

    histiocytes, intermediate cells and interdigitating cells

    of a dendritic cell lineage, T-cell lymphocytes, eosin-

    ophils, and macrophages. The hallmark cell is the

    Langerhans cell histiocyte. This cell has abundant

    eosinophilic to amphophilic cytoplasm and a nucleusthat appears reniform, deeply indented, or grooved. The

    number of eosinophils is quite variable from being

    abundant with eosinophilic abscesses to sparse or even

    absent. Occasional giant cells representing fusion of

    either macrophages or Langerhans cell histiocytes may

    be seen. The presence of this granulomatous inflamma-

    tion with occasional giant cells raises the concern for an

    infectious process, such as tuberculosis in the past, and

    viral infection with an agent that is capable of inducing

    syncytial (giant) cells. Necrosis within this granulom-

    atous lesion is not unusual, and again reinforced the

    suggestion in the past that these lesions represented aninfectious process. The lesions vary from an indistinct

    focus with blending into the adjacent normal tissue to

    nodular in appearance, depending on tissue types in-

    volved. Although present, mitotic activity tends to be

    low to moderate without atypical mitotic figures. The

    lesions may take on a more atypical appearance and

    appear as epithelioid granulomas (Fig 5, D) that lack

    the typical features of LCH. The lesions may resemble

    other histiocytic lesions, such as early juvenile xantho-

    granuloma that lack characteristic Touton giant cells.

    Table V. Comparison of childhood and adult Langer-hans cell histiocytosis

    Children Adults

    Bone disease Extremely frequent Frequent

    Jaws 8% 30%

    Skull 40% 21%

    Skin disease Frequent Frequent

    Dental disease Infrequent Frequent

    Pulmonary disease Infrequent Very frequent

    Pulmonary disease alone Rare Very frequent

    Genital involvement Rare Frequent

    Diabetes insipidus Frequent Frequent

    Compiled from references 22-24.

    OOOOE

    S48 Hicks and Flaitz August 2005

  • 7/21/2019 LCH Sunil Sir

    8/25

    Definitive diagnosis with these atypical lesions requires

    immunocytochemistry and occasionally electron mi-

    croscopy.

    Immunocytochemical featuresLCH can be distinguished from other dendritic cell

    disorders on the basis of cytoplasmic and cell surface

    markers that it expresses (Table VII, Fig 6).5,11,12,21,36-38

    In the past, S100 protein, Lag antigen, and peanut

    agglutinin were used. With the development of more

    specific and sensitive antibodies to cell surface mark-

    ers associated with Langerhans cell histiocytes, these

    markers are rarely used for diagnostic purposes. CD1a

    (OKT6) is a well-recognized marker that immunoreacts

    with Langerhans cells in the epidermis. This antibody

    also identifies Langerhans cell histiocytes, cortical

    thymocytes, and interdigitating dendritic cells within

    the dermis andlymph nodes. Langerhans cell histiocytes

    Fig 1. Clinical appearance of oral and maxillofacial Langerhans cell histiocytosis (LCH).A, Seborrheic-like dermatitis appearance

    of LCH in postauricular region.B, Diffuse gingival enlargement secondary to LCH. C, Infiltration of maxillary frenum by LCH.

    D, Maxillary alveolar and palatal LCH mass. E, Palatal diffusely ulcerated LCH. (Fig 1, A and B courtesy of Dr Moise Levy,

    Houston Tex; Fig 1C courtesy of Dr Megan Dishop, Houston Tex; Fig 1, E reprinted with permission from Eisen D, Lynch D,

    editors. The mouth: diagnosis and treatment. St. Louis, Mo: Elsevier Publishers; 1998.)

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S49

  • 7/21/2019 LCH Sunil Sir

    9/25

    demonstrate a membranous to cytoplasmic pattern

    with the CD1a antibody (Fig 6, A). According to the

    Histiocyte Society, a definitive diagnosis of LCH maybe rendered with the appropriate histopathology and

    when immunoreaction with CD1a is present. This

    antibody may be used with formalin-fixed paraffin-

    embedded tissues, and eliminates the need for frozen

    tissue for immunocytochemistry.

    More recently, a highly specific and sensitive anti-

    body against Langerhans cells and Langerhans cell

    histiocytes has become commercially available (Fig 6,

    B).21,39-43 CD207 (langerin) is a monoclonal antibody

    against a type II transmembrane protein expressed with

    Langerhans cells and Langerhans cell histiocytes.

    CD207 reacts with a 40-kD langerin protein purportedly

    specific to these cells. Langerin is located on the cellsurface of Langerhans cells and Langerhans cell histi-

    ocytes and induces membrane superimposition and

    zippering that leads to Birbeck granule formation

    (LC granules, Birbeck-Broadbent granules). These

    pentalaminar structures with a bulbous end (tennis

    racket and handle) represent a means to capture antigen

    for antigen processing. These structures are rapidly

    internalized into the cells. The CD207 (langerin) anti-

    body requires antigen unmasking with formalin-fixed,

    paraffin-embedded tissues.

    Fig 2. Diagnostic imaging of skull and rib in LCH. A, Well-circumscribed parietal bone LCH lesion. B, Destruction of

    right zygomatic process of maxilla secondary to LCH with significant soft tissue component. C, Bilateral mastoid involvement by

    LCH with soft tissue mass anteriorly displacing ears bilaterally. D, Left rib with loss of cortical bone and soft tissue mass bulging

    into parietal pleura.

    OOOOE

    S50 Hicks and Flaitz August 2005

  • 7/21/2019 LCH Sunil Sir

    10/25

    Fig 3. Clinical and radiographic appearance of oral and maxillofacial LCH.A and B, Young child with gingival soft tissue LCH

    mass (A) with mild lingual displacement of the primary molars and a diffuse radiolucent lesion ( B) with loss of bone surrounding

    the primary first molar and mesial root of primary second molar. C-E,A 34-year-old woman with erythematous lingual mucosa

    adjacent to a second molar (C) with root canal therapy 6 years previously. Periapical radiograph (D) with diffuse bone loss

    secondary to biopsy-proven LCH. E, Same patient with progressive mandibular and maxillary LCH involvement with tooth loss

    over a 3-year period. F and G, Elderly man with diffusely erythematous mandibular gingiva and mucosa with tissue loss due to

    LCH (F). Periapical radiograph (G) reveals diffuse bone loss secondary to LCH. (Fig 3, A and B reprinted with permission from

    Neville B, Damm D, Allen C, Bouquot J, editors. Oral and maxillofacial pathology. 2nd ed. St. Louis, Mo: WB Saunders Co; 2002.

    Fig 3, C, D, and Ecourtesy of Dr Brad Neville and Dr John Hann, Charleston SC. Fig 3, Fand G courtesy of Dr Alan Gould,

    Crestwood, Ky.)

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S51

  • 7/21/2019 LCH Sunil Sir

    11/25

    Fig 3. Continued.

    OOOOE

    S52 Hicks and Flaitz August 2005

  • 7/21/2019 LCH Sunil Sir

    12/25

    Fig 4. Congenital self-healing LCH.A, Periorbital nodular lesions in a neonate. B, Characteristic histopathologic appearance of

    LCH with admixture of Langerhans cell histiocytes with deeply indented, grooved, and reniform nuclei and abundant eosinophilic

    to amphophilic cytoplasm, eosinophils and lymphocytes (hematoxylin and eosin, original magnification 3400). C, Concentric

    laminated bodies in Langerhans cell histiocytes pathognomonic for congenital self-healing LCH (transmission electron

    microscopy, original magnification 310 000). (Fig 4, A courtesy of Dr Denise Walker Metry, Houston Tex.)

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S53

  • 7/21/2019 LCH Sunil Sir

    13/25

    Differentiation of LCH from other dendritic cell dis-

    orders is important. The immunophenotypes of dendritic

    cell disorders are presented in Table VII.5,11,12,21,36-43

    Dendritic cell disorders may contain a certain percent-

    age of normal Langerhans cells that are trafficking

    back to regional lymph nodes to perform their antigen-

    presenting cell functions. In such cases, where there may

    be a certain population of Langerhans cells or macro-

    phages that have morphology similar to Langerhans cell

    histiocytes, further characterization with immunocyto-

    chemistry would be necessary. Xanthogranulomatous

    processes are defined by immunoreaction with dendritic

    cell markers (Factor XIIIa, fascin) and macrophage

    markers (CD68 [PGM1], CD163). It is important to

    recognize that juvenilexanthogranuloma may be seen

    in children with LCH.44-46 Juvenile xanthogranuloma

    may appear before, concurrent with, or following the

    diagnosis of LCH. Children with juvenile xanthogran-

    ulomas should be evaluated for and have appropriate

    follow-up to assess concurrent or development of LCH.

    Rosai-Dorfman disease is characterized by reactivity

    with S100 protein and macrophage markers, as well as

    Table VI. Oral and maxillofacial Langerhans cellhistiocytosis: clinical features*

    Mean age, y (range) 18.9 (Neonate-53)

    Age, y

    \10 29%

    10-19 22%

    20-39 44%

    40-53 5%Gender ratio, M:F 3.9:1.0

    Unifocal disease, % 49

    Multifocal single system disease, % 25

    Multifocal multisystem disease, % 25

    Sites involved, %

    Jaws 67

    Mandible 76

    Posterior 68

    Anterior 32

    Maxilla 24

    Posterior 63

    Anterior 37

    Oral soft tissues 33

    Gingiva 76

    Hard palate 10

    Floor of mouth 6

    Maxillary sinus 5

    Buccal mucosa 4

    Symptoms at presentation, %

    Mass/Lesion 59

    Pain 46

    Gingivitis 43

    Loose teeth 41

    Oral ulcer 19

    Impaired healing 13

    Halitosis 12

    Masticatory problems 9

    Vincents infection 7

    Gingival bleeding 6Paresthesia 4

    Extraoral symptoms at presentation, % 70

    Extraoral lesions 50

    Soft tissue lesion only 7

    Bone and soft tissue 43

    Diabetes insipidus 16

    Lymphadenopathy 16

    Anemia 15

    Skin lesions 12

    Exophthalmos 10

    Hepatosplenomegaly 9

    Otitis media 8

    Fever 7

    Leukocytosis 2

    Mental disability 2Extraoral sites of involvement, %

    Skull 37

    Lower extremity 28

    Upper extremity 13

    Ribs 13

    Lung 12

    Skin 10

    Liver 10

    Spine 7

    Orbit 7

    Cervical lymph nodes 6

    Table VI. Continued

    Submandibular gland 2

    Parotid gland 2

    Radiographic appearance, %

    Unilocular radiolucent lesions 100

    Well-demarcated borders 63

    Borders poorly corticated 24

    Poorly defined borders 12Displaced teeth 51

    Root resorption 17

    Periosteal reaction 8

    Treatment, %

    Surgery alone 50

    Radiation therapy alone 20

    Surgery and radiation therapy 23

    Chemotherapy 7

    Intralesional steroids 1

    Recurrence, %

    Unifocal disease, 16

    Multifocal single system disease 22

    Multifocal multisystem disease 15

    Single recurrence 15

    Two recurrences 4

    Surgery alone 12

    Radiation therapy 25

    Surgery and radiation therapy 19

    Chemotherapy 0

    Outcome, %

    No evidence of disease 53

    Alive with disease 17

    Alive, status not known 7

    Died of disease 7

    Died of other causes 11

    Died of unknown cause 3

    *Compiled from references27-35.

    OOOOE

    S54 Hicks and Flaitz August 2005

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    14/25

    emperipolesis (leukocytes passing through the cyto-

    plasm of macrophages).5-11,12,21,36-43 Indeterminantdendritic cell histiocytomas immunoreact with CD1a,

    S100 protein, and fascin.5-11,12,21,36-43 Interdigitating

    dendritic cell histiocytoma have a similarimmunophe-

    notype, but also display CD83 expression.5-11,12,21,36-43

    Follicular dendritic cell histiocytoma becomes an issue

    if a lymph node or the spleen contains a histiocytic

    lesion.5-11,12,21,36-43

    The expression of CD21 and CD35would help to differentiate this tumor from other

    histiocytic lesions.

    Ultrastructural featuresThe gold standard for definitive diagnosis of LCH

    is examination of tissue using transmission electron

    microscopy (Table VIII, Fig 7).1,2,4-6,9,11,12,21,38 The

    identification of Birbeck granules in the cytoplasm or

    within the cell border of lesional cells provides the final

    evidence necessary in difficult to characterize tumors, or

    when there are aberrant immunocytochemical results.

    All forms of LCH possess Birbeck granules. The typicalgranule is composed of a rod-shaped or tennis rackete

    shaped structure with a length varying from 200 to

    400 nm and a width of 33 nm. The rod (handle) region

    has a zipper-like morphology and has been described as

    pentalaminar owing to a central striated membrane anda

    double electron dense outer sheath. The granules are

    more easily identified in early lesions, rather than inlong-standing lesions. Birbeck granules tend to be rare

    when LCH involves the liver, spleen, and gastrointesti-

    nal tract. Other ultrastructural structures present within

    the cytoplasm of Langerhans cell histiocytes are curvi-

    linear and multivesicular bodies, tubuloreticular inclu-

    sions, and cylindrical confronting cisterna.

    Congenital self-healing LCHCongenital self-healing LCH deserves special atten-

    tion because this disease spontaneously resolves over

    Fig 5. Histopathologic features of LCH. A, Abundant eosinophils with frequent histiocytes and less apparent lymphocytes

    (hematoxylin and esocin, original magnification 3100). B and C, Langerhans cell histiocytes with abundant eosinophilic to

    amphophilic cytoplasm and deeply indented, reniform, and grooved nuclei (hematoxylin and eosin, original magnification 3400).

    D,Atypical LCH lesion composed of epithelioid Langerhans cell histiocytes with abundant eosinophilic cytoplasm and lacking

    typical reniform nuclei (hematoxylin and eosin, original magnification 3200). These histiocytic cells immunoreacted with CD1aand CD207, as well as demonstrated Birbeck granules (not shown).

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S55

  • 7/21/2019 LCH Sunil Sir

    15/25

    several months and may be confused with multifocal

    single system or multifocalmultisystem disease that

    requires treatment.4,17,21,25 Congenital self-healing

    LCH has an identical histopathology and immunophe-

    notype like all other forms of LCH, but has unique

    ultrastructural features (Fig 4, Table VIII). Transmissionelectron microscopy will reveal laminated dense bodies

    with a concentric myelin-like morphology and non-

    laminated dense bodies. Birbeck granules are found, but

    tend to be detected in less than one third of the

    Langerhans cell histiocytes. The presence of laminated

    dense bodies provides definitive proof for the diagnosis

    of congenital-self healing LCH. This form of LCH

    occurs predominantly in children younger than 1 year of

    age. It is recommended that any child younger than 2

    years with evidence of LCH should have transmission

    electron microscopy performed on the biopsy or resec-

    tion tissue to confirm or refute the diagnosis of typical

    LCH or congenital self-healing LCH.

    Tissue availability for electron microscopicevaluation

    Ideally, tissue should be set aside in 2% to 3% bu f-

    fered glutaraldehyde for possible ultrastructural study.21

    In most oral and maxillofacial pathology services,

    electron microscopy is not a high priority. Ultrastruc-

    tural examination can be performed on residual tissue

    fixed in buffered 10% formalin without deleterious

    results. Alternatively, a portion of lesional tissue may be

    removed from a tissue block and undergo paraffin

    recovery, and then transmission electron microscopic

    examination. Ultrastructure may be compromised to a

    certain extent, however the tumor-defining structures

    (Birbeck granules, laminated dense bodies) are usually

    of adequate quality to provide for a definitive diagnosis.Stringent control of processing temperature of the tissue

    and low-melting point paraffin for embedding the tissue

    allows for better preservation of ultrastructural features.

    RECOMMENDED EVALUATION FOLLOWINGDIAGNOSIS OF LANGERHANS CELLHISTIOCYTOSIS

    Once a biopsy-proven diagnosis is obtained, the

    patient should undergo referral to a specialist who is

    well versed in LCH (pediatric or adult hematologist/

    oncologist). For appropriate classification, the following

    are recommended4,6,9,17:

    d Thorough clinical examinationd Complete blood cell count with hematocrit, hemo-

    globin, and platelet countd Liver function tests (serum transaminases, alkaline

    phosphatase, bilirubin, albumin, total protein)d Coagulation studies (prothrombin and partial throm-

    boplastin time, fibrinogen)d Urine osmolarityd Arginine vasopressin study for diabetes insipidusd Chest radiographd Skeletal radiograph surveyd

    CT or ultrasound of chest and/or abdomen if organsystem involvement suspectedd MRI of the brain if central nervous system involve-

    ment suspectedd Respiratory function tests if respiratory involvement

    suspectedd Diagnostic endoscopy if gastrointestinal symptoms

    suspected

    BIOLOGIC, CYTOGENETIC, ANDMOLECULAR ASPECTSLCH: The Chemokine/Cytokine Storm

    As noted previously, Langerhans cell histiocytes areconsidered to be immature dendritic cells that fail to

    undergodifferentiationto mature dendritic cells.4,15,47-49

    Similar to immature dendritic cells, Langerhans cell

    histiocytes express receptors for inflammatory chemo-

    kines (CCR1, CCR2, CCR5, CCR6, CXCR1), which

    allow for the recruitment of additional immature den-

    dritic cells (Table IX).50-54 Langerhans cell histiocytes

    fail to mature and do not down-regulate the above-

    mentioned chemokines associated with immature den-

    dritic cells. In addition, these histiocytes fail to display

    Table VII. Immunocytochemical features of dendriticcell disorders*

    Langerhans cell histiocytosis

    Markers important for diagnosis:

    CD1a, CD207 (Langerin), S100, Lag antigen

    Additional markers:

    HLA-DR, E-cadherin, peanut agglutinin, CD4, CD31, CD40,

    CD49d, CD52, CD54, CD80, CD86, CD116 (GM-CSFR),CD209 (DC-SIGN), CCR6, PLAP, NSE, vimentin, IL2-R

    (CD25), IFN-gamma, TNF-alpha, acid phosphatase, CD68

    (weak), LCA (CD45 weak), lysozyme weak)

    Xanthogranuloma family (juvenile xanthogranuloma,

    Erdheim-Chester disease, xanthoma disseminatum,

    dermal dendrocytomas)

    Factor XIIIa, Fascin, CD68 (PGM1), CD163, CD14,

    Ki-M1P, CD45

    Rosai-Dorfman disease (sinus histiocytosis with massive

    lymphadenopathy, sinus dendritic cell)

    CD68, S100, fascin, CD163, cathepsin E, alpha-1-antitrypsin,

    Si-M9, CD31

    Dendritic cell histiocytoma, indeterminant cell type

    CD1a, S100, fascin, CD45

    Dendritic cell histiocytoma, interdigitating dendritic cell type

    CD1a, S100, Fascin, CD83, CD45

    Dendritic cell histiocytoma, follicular dendritic cell type

    CD21, CD35, Ki-M4, Fascin, S100 variable (6)

    *Compiled from references5, 11, 12, 21, 36-43.

    OOOOE

    S56 Hicks and Flaitz August 2005

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    16/25

    chemokines (CXCR4, CCR7) that are constitutively

    expressed by mature dendritic cells. High levels of

    other inflammatory chemokines (CCL20/MIP-3alpha,

    CCL2/MCP-1, CCL3/MIP-1alpha, CCL4/MCP-4alpha,

    CCL5/RANTES, CXCL8/IL-8, CXCL10/IL-10) are re-

    leased by Langerhans cell histiocytes, and these chemo-

    kines are also associated with immature dendritic cells.

    These chemokines are important in the recruitment of

    circulating immature dendritic cells, as well other im-

    mune cell types (T lymphocytes, macropaghes, eosino-

    phils). Langerhans cell histiocytes have high levels of

    CD14, CD86, and intracellular MHC class II, and lack

    CD83, CD86, and DC-LAMP, which are characteristicfor immature dendritic cells.4-15,17 It is quite obvious why

    Langerhans cell histiocytes are considered to be in an

    arrested state of activation and differentiation. This would

    also explain thelack of thin, fingerlikedendritic processes

    seen in normal Langerhans cells (mature dendritic cells),

    and the characteristic ovoid to round morphology of

    Langerhans cell histiocytes (immature dendritic cells).

    The lack of maturation also prevents Langerhans cell

    histiocytes from leaving the lesion site.

    Up-regulation of many cytokines have been demon-

    strated with LCH (Table IX).17,52-60 Histopathologic

    examination of lesions notes a mixture of Langerhanscell histiocytes, lymphocytes (predominantly helper

    [CD4] T lymphocytes), eosinophils, and macrophages.

    Each of these cell types produces cytokines in the

    lesions, which result in autocrine and paracrine stimu-

    lation of the lesional cells. The major players are con-

    sidered to be the Langerhans cell histiocytes and T-cell

    lymphocytes (predominantly helper CD4 lymphocytes).

    Major factors that participate are the cytokines IL-1,

    IL-3, IL-4, IL-5, IL-8, IL-10, GM-CSF, TNF-alpha,

    transforming growth factor (TGF)-beta, and leukemia

    inhibitory factor (LIF) (Table IX). Many of the cyto-

    kines favor recruitment of Langerhans cell progenitors

    and rescue from apoptosis. Langerhans cell histiocytes,

    macrophages, and T cells produce IL-10, GM-CSF, and

    interferon (INF)-alpha. IL-1 is derived from Langerhans

    cell histiocytes. T-cell lymphocytes synthesize IL-2,

    IL4, IL-5 and TNF-alpha. Eosinophils produce IL-5,

    INF-alpha, GM-CSF, IL-7, and IL-10.

    These cytokines are important because they create

    a cytokine storm in which autocrine and paracrine

    stimulation is established among the cells, and in

    particular, the Langerhans cell histiocytes and helper

    T lymphocytes (CD4).17,52-60 For example, Langerhanscell histiocytes are stimulated by IL-1 and IL-2 pro-

    duced by T lymphocytes. Both of these cell types also

    upregulate INF-alpha, which stimulates IL-1 produc-

    tion. Recruitment of Langerhans cell progenitors

    occurs, in part, under the influence of GM-CSF that

    is produced by both Langerhans cell histiocytes and

    CD4 T lymphocytes. Interestingly, serum GM-CSF and

    IL-2 levels are dramatically increased in patients

    with multifocal multisystem disease. GM-CSF, IL-3,

    TNF-alpha and other cytokines act as chemoattractants

    for CD341 Langerhans cell precursors, eosinophils,

    neutrophils, and macrophages. TNF-alpha inhibitsspon-taneous apoptosis of Langerhans cell histiocytes, there-

    by promoting their survival. This anti-apoptotic effect is

    further enhanced by IL-1-alpha, GM-CSF, and IL-3.

    Only recently has it been possible to evaluate

    expression profiles for LCH using laser capture micro-

    dissection, RNA amplification, and gene microarray

    techniques.60 Certain differences in cytokine expression

    have been found. With LCH involving the liver and

    spleen, the cytokines IL-13, nuclear factor kappa

    B (NFkB), TNF receptor, p55, receptor-activator of

    Fig 6. Immunocytochemical identification of LCH. A, Langerhans cell histiocytes reacting in a cytoplasmic pattern with

    CD1a (immunocytochemistry anti-CD1a antibody, original magnification 3400). B, Langerhans cell histiocytes reacting in a

    cytoplasmic pattern with CD207/langerin (immunocytochemistry anti-CD207 antibody, original magnification 3400).

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S57

  • 7/21/2019 LCH Sunil Sir

    17/25

    NF-kappa B ligand (RANKL), CD40-L and TGF-beta

    receptor are significantly upregulated, compared with

    normal Langerhans cells. Bone-only disease has a

    different expression profile than multisystem disease.

    The upregulated cytokines and genes in bone-only

    disease, compared with multisystem disease, are CD40/

    CD40L, GM-CSF, GM-CSF receptor, IL-1-3 receptor-1,

    IL-2, IL-5 receptor, TNF-alpha, TGF-beta, TNF re-

    ceptor associated factor 1 (TRAF1), and TRAF6. The

    discovery of differences in cytokine and gene profile

    expression for specific forms of LCH provides potential

    avenues to explore in understanding pathogenesis andnew targets for therapy.

    The symptoms associated with LCH, such as fever

    and failure to thrive, have been associated with the

    chemokine/cytokine storm effect.17,50-57,60 In addi-

    tion, several of these factors promote osteoclastic

    activity and osteolysis, promote fibrosis (liver andlung fibrosis, hypothalamic/pituitary axis sclerosis

    leading to diabetes insipidus), hematologic dysfunction,

    angiogenesis, and overgrowth of granulation tissue.

    In addition to the chemokines and cytokines involved

    in lesion formation with LCH, many different growth

    factors, regulators of the cell cycle, adhesion molecules

    andimmunologicmarkers have been implicated (Table

    IX).17,52,53,56,58-69 It is obvious that the process of lesion

    formation in LCH is quite complex, and involves

    numerous mediators that cross-talk among the vari-ous cell types.

    Lack of viral etiologyAt one point, the issue of whether LCH had a viral

    etiology was raised. Convincing evidence from several

    investigations using in situ hybridization and poly-

    merase chain reaction techniques, as well as viralcul-

    tures, has not surfaced to support a viral etiology.69-71

    Adenovirus, cytomegalovirus, Epstein-Barr virus, her-

    pes simplex virus, human herpesvirus type 6, HIV,

    human T-cell leukemia virus types I and II, and

    parvovirus have been shown not to be present in LCH.Some investigators have noted immunohistochemicalstaining for human herpesvirus 6 in lesions, but there

    were no serologic data that showed a recent infection

    had occurred. It was concluded that reactivation of a

    latent viral infection could be the explanation, which is

    common among herpesvirus family members.

    Neoplastic process, cytogenetics, and moleculargenetics in LCH

    During the past decade, there has been controversy

    regarding whether LCH is a neoplastic or reactive

    process (Table X).17,47,48,72-74 Clonal expansion of

    Langerhans cell histiocytes has been shown repeatedly

    in eosinophilic granuloma, Letterer-Siwe disease, and

    Hand-Schuller-Christian disease. Clonality in these

    disease processes has been proven by both human

    androgen receptor (HUMARA) DNA assay and T-cell

    receptor analysis.17,72-74 Of interest is the fact that the

    pulmonary only form of LCH that occurs in young adult

    smokers has not proven in multiple investigations to be

    clonal. This would fit the clinical scenario, because it is

    possible for these lesions to regress with smoking

    cessation. Although not studied, lack of clonality in

    congenital self-healing LCH would be expected.

    Further evidence for a neoplastic process is familialclustering of LCH with multiple affected rela-

    tives.17,74,75 Familial clustering of LCH accounts for

    only about 1% of the cases. An association between

    Table VIII. Langerhans cell histiocytosis: diagnosticcriteria

    Presumptive diagnosis

    Granulomatous inflammation and hallmark Langerhans cell

    histiocytes identified on routinely stained tissue sections

    (characteristic histopathology)

    Definitive diagnosis

    Characteristic histopathology and Birbeck granules on electronmicroscopy and/or CD1a immunoreactivity (CD207

    Langerin substitute for CD1a)

    Immunocytochemistry important for diagnosis

    Positive for: CD1a, CD207 (langerin), S100 protein,

    peanut agglutinin (PNA)

    Negative for: Factor XIIIa, fascin

    Ultrastructural features important for diagnosis

    Langerhans cell histiocytosis

    LC granules (Birbeck granules, pentalaminar structures)y

    Multivesicular bodies

    Curvilinear membranous formations (worm structures)

    Tubuloreticular structures

    Cylindrical confronting cisternae (comma-shaped structures)

    Congenital self-healing Langerhans cell histiocytosis

    (Reticulohistiocytosis; Hashimoto-Pritzker disease)

    Laminated dense bodies (concentric, myelin-like bodies)y

    Nonlaminated dense bodiesy

    LC granules (Birbeck granules, pentalaminar structures,

    \30% of Langerhans cell histiocytes)y

    Compiled from references 1-6, 9-12, 17, 21, 25, 38.yTumor-defining structures.

    Fig 7. Ultrastructure of LCH.A, Large histiocytic cells with abundant cytoplasm and deeply indented and irregular nuclei with

    dispersed chromatin (transmission electron microscopy, original magnification 31500).B and C, Birbeck granules (LC granules)

    characterized as tennis rackets with handles (arrow). More often, Birbeck granules are found as pentalaminar rod (handle only of

    tennis racket) structures only, with some having rudimentary bulbous ends (transmission electron microscopy, original

    magnification 328 000). (Fig 7, B and Ccourtesy of Dr Gary Mierau, Denver, Colo.)

    OOOOE

    S58 Hicks and Flaitz August 2005

  • 7/21/2019 LCH Sunil Sir

    18/25

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S59

  • 7/21/2019 LCH Sunil Sir

    19/25

    mannose-binding lectin alleles and susceptibility to

    LCH in family members has been suggested as one

    possible mechanism. In addition, LCH has occurred in

    identical twins, with similar clinical features, dissem-

    inated disease, and same sites of involvement.74,75 There

    is an 86% concordance rate for identical twins, but no

    evidence for concordance with fraternal twins.

    Certain HLA haplotypes in different ethnic groups

    increase the susceptibility for development of LCH.76

    The exact reason for this is not known currently, but is

    under investigation. Constitutional or acquired chromo-

    somal instability may be another factor in LCH

    development.74,75 This is illustrated by a child with an

    unknown DNA breakage syndrome who developed

    LCH, in addition to ataxia, ocular telangiectasia, and

    chromosomal instability.77 Finally, there is a known

    increased incidence of other malignancies in children

    with LCH (acute and chronic leukemias, lymphomas,

    solid tumors;Table II).4,17,26,69,74,78

    Comparative genomic hybridization (CGH), loss of

    heterozygosity (LOH), and conventional cytogenetics

    have demonstrated several genomic abnormalities intissue from LCH (Table X).73,79-81 In particular, certain

    chromosomal sites that have losses or gains in DNA

    sequences and/or loss of heterozygosity in LCH are loci

    where tumor suppressor genes are housed. One such

    site in LCH is the short arm (p) of chromosome 1,

    particularly 1p36. There are several tumor suppressor

    genes at the 1p36 locus (IDE, CDC2L1, PAN, PAX7,

    E2F2,TNFR-2,TCEB). Deletions and loss of heterozy-

    gosity of the 1p arm have also been associated with

    neuroblastoma, B cell chronic lymphocytic leukemia,

    and Wilms tumor. Loss of chromosome 7 has been

    noted on CGH and LOH studies with LCH. The tumorsuppressor gene plasminogen activator inhibitor is

    located on this chromosome. Chromosome 7 loss and

    abnormalities are linked to Fanconis anemia, myelo-

    dysplastic disorders, and acute myeloid leukemia. 49,78

    With LCH, chromosome 9 is affected by loss of DNA

    sequences and loss of heterozygosity at 9p21.73,79-81

    At chromosome 9p21 are several regulators of the

    cell cycle (cyclin-dependent kinases, cyclin-dependent

    kinase inhibitors, CDKN2A, p14, p15, p16).

    Chromosome 9q is where the tuberous sclerosis tumor

    Table IX. Cell proliferation factors in Langerhans cellhistiocytosis

    Chemokines

    CCR1

    CCR2

    CCR5

    CCR6

    CCL2/MCP-1CCL3/MIP-1alpha

    CCL4/MCP-4alpha

    CCL5/RANTES

    CCL17/TARC

    CCL20/MIP-3 alpha

    CXCL8/IL-8

    CXCL10/IL-10

    CXCL11/I-TAC

    CXCR1

    CXCR3

    Cytokines

    Interleukins (IL) 1, 2, 3, 4, 6, 8, 9, 10, 11, 12, 13, 17, 22

    Interferon-gamma (IFN)

    Tumor necrosis factor (TNF)-alpha

    TNF-receptor (p75)

    TNF receptor associated factor (TRAF1, TRAF6)

    Granulocyte macrophage-colony stimulating factor (GM-CSF)

    GM-CSF receptor (GM-CSFR)

    Macrophage-colony stimulating factor (M-CSF)

    Stem cell factor (CD341)

    Leukemia inhibitory factor (LIF)

    CD40 ligand and receptor

    Transforming growth factor (TGF)-alpha and beta

    TGF-beta receptor

    RANKL (receptor-activator of NF-kappa B ligand)

    NF-kappa B (nuclear factor kappa B)

    FMS-like tyrosine kinase ligase (FLT3-L; colony stimulating

    factor 1 receptor, formerly McDonough feline sarcoma viral

    oncogene homolog)Angiotensin-converting enzyme (ACE)

    Prostaglandin E2

    Exotaxin 1, 2, 3

    Leptin

    Growth Factors

    Fibroblastic growth factor 6, 14 (FGF)

    Vascular endothelial growth factor (VEGF)

    Platelet-derived growth factor-receptor beta (PDGF-R beta)

    PDGF-alpha

    Bone morphogenic protein 8 (BMP)

    Thrombopoietin

    Allograft inflammatory factor 1

    Cell cyclerelated gene products

    MDM2

    p53

    p55

    p21

    p16

    Rb

    Bcl2

    Cell adhesion

    CD2

    CD44

    CD54

    CD58

    CD11c

    Table IX. Continued

    Immunologic markers

    CD1a (OKT6)

    CD207 (langerin)

    CD4

    CD45

    MHC II

    Compiled from references4-15, 17, 47-69.

    OOOOE

    S60 Hicks and Flaitz August 2005

  • 7/21/2019 LCH Sunil Sir

    20/25

    suppressor gene is found. Loss of heterozygosity with

    the short arm (p) of chromosome 17 has been identified

    in LCH.73,79-81 The tumor suppressor genep53 is located

    at 17p13. Loss of heterozygosity has been demonstrated

    on the long arm (q) of chromosome 22 in LCH. 73,79-81

    The tumor suppressor gene NF2 is located on chromo-

    some 22q. In addition, gains at chromosome 4q and 12phave been observed in osteosarcomas (4q), gliomas

    (12p), and B-cell chronic lymphocytic leukemias

    (12p).73,79-81 LCH has been shown to have DNA

    sequence gains along the long arm (q) of chromosome

    4 and with chromosome 12.

    The finding of cytogenetic abnormalities in chromo-

    some 7 (Table X) has further strengthened the link

    that LCH is a myeloid stem cell disorder.49,73,78-81

    Chromosomal abnormalities associated with chromo-

    some 7 include Fanconis anemia, myelodysplastic

    disorders, and acute myeloid leukemia. Recently, there

    have been several children with concurrent LCH and

    myelodysplasia. Interestingly, myelofibrosis may be

    seen in LCH and has been ascribed to bone marrow

    involvement. Perhaps greater attention will be directedtoward chromosome 7 in LCH on the basis of these

    findings.

    Cytogenetic studies (Table X) have shown that

    tumor suppressor genes (p53), oncogenes (c-myc,

    h-ras), adhesion molecules (E-cadherin), cell surface

    immunologic markers, growth factors, and apoptotic

    defects participate in LCH to some degree.69,72-75,80,81

    These factors represent potential targets for therapy.

    CURRENT TREATMENT PROTOCOLS ANDINNOVATIVE THERAPEUTIC AGENTS

    Over the past decade, the Histiocyte Society initiated

    a series of treatment protocols for LCH aimed at the

    pediatric population, andmore recently has opened an

    adult protocol (Table XI).15,17,82-86 The first pediatric

    protocol (LCH-I) began in 1991 and was targeted

    toward multisystem disease. The study analyzed the

    effect of vinblastine and etoposide in combination with

    intravenous steroids. The overall response was similar

    with either treatment (58% vs 65% for vinblastine vs

    etoposide). Survival was also not statistically different

    between the groups (76% for vinblastine, 83% foretoposide). Of interest was the determination that chil-

    dren who failed to respond to therapy within a 6-week

    period were at increased risk for treatment failure and

    required a different therapy. Mortality in these poor

    responders was 66%. LCH-II compared treatment with

    vinblastine, oral prednisone, and mercaptopurine with

    or without etoposide. The addition of etoposide did not

    provide any difference in survival, relapse, or toxicity.

    Because of the risk for a second malignancy (myeloid

    leukemia) with etoposide, this drug is no longer used

    in LCH protocols. The prior finding of decreased sur-

    vival in children who did not respond to therapy after

    6 weeks was again noted (good responders 88% sur-

    vival, poor responders 17% survival). The current

    protocol (LCH-III) compares different therapies for

    multisystem disease with high-risk sites, multisystem

    disease with low-risk sites, multifocal single system

    disease, and localized special site disease. This study

    is currently accruing patients and results are not yetavailable.

    The Histiocyte Society has recently launched a

    protocol for adults with LCH (Table X).17,82-86 The

    treatment groups are single system disease, central

    nervous system disease, isolated pulmonary disease,

    and multisystem disease. Accrual is ongoing and results

    are not available at the present time.

    The therapies available for those children and adults

    not enrolled into the Histiocyte Society protocols are

    determined by the patients oncologist on a case-by-case

    Table X. Langerhans cell histiocytosis: cytogeneticsand molecular genetics*

    Evidence for neoplastic process

    Clonal expansion of Langerhans cell histiocytosis

    Human androgen receptor (HUMARA) DNA assay

    T-cell receptor analysis

    Familial occurrence with multiple affected relatives and

    identical twinsMannose-binding lectin allele

    Immune system gene dysregulation

    Increased incidence of other malignancies

    HLA haplotypes (chromosome 6)

    DR4, Cw7, B7, B8 (Caucasians, United States)

    B7, DR2 (England)

    B61, Cw7 (Japanese)

    Comparative genomic hybridization (CGH)

    Loss of DNA sequences on chromosomes

    1p (1p36, 1p21-36), 2, 5, 6, 7, 9, 11p, 16, 17, 22q

    Gain of DNA sequences on chromosomes

    2q, 4q, 12

    Loss of heterozygosity (LOH) with chromosomes

    1p, 1p36, 1p22, 1p13, 1q31, 1q, 3p, 4q35, 5q, 7, 7p21, 7p15, 9, 9p,

    9p21, 17p, 22, 22q, 22q11

    Other cytogenetic abnormalities reported

    t(7;12)(q11.2;p13)

    Myelodysplastic disorder association (chromosome 7)

    Paracentric inversion 13q

    Susceptibility to chromatid and chromosomal breakage

    Constitutional chromosomal instability

    Chromosomal pulverization with herpesvirus and respiratory

    syncytial virus

    p53 overexpression

    c-myc overexpression

    H-ras overexpression

    E-cadherin underexpression

    Aneuploidy (DNA index[1.5)

    Apoptosis defects

    *Compiled from references17, 21, 69,72-75, 80, 81.

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S61

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    21/25

    basis.17,82-85 Therapies vary considerably from surgical

    curettage to excision with or without the addition of

    radiation or chemotherapy. The Histiocyte Society is

    contemplating recommending multiagent chemother-

    apy for bone-only disease (unifocal or multifocal). Thisis based on clinical data indicating that single agent

    chemotherapy, radiation therapy, or observation only is

    associated with a high recurrence rate (50% to 80%)

    compared with multiagent chemotherapy (18%, Table

    X). In fact, some knowledgeable clinicians recommend

    chemotherapy with extensive jawinvolvement to avoid

    heroic surgery and loss of teeth.17 They contend that a

    6-month course of vinblastine and prednisone has a high

    likelihood to cure the disease, allow bone redeposition,

    and avoid tooth loss.

    Certain late effects among pediatric survivors of

    LCH are more common that others (Table X).15,17,82-86

    Late effects are about 3 times as common with multi-

    system disease than with single system disease. Diabetes

    insipidusand orthopedic problems occurin about 20%to

    25% of children. Tooth loss is noted in less than 10%.

    INNOVATIVE THERAPEUTIC AGENTSWith the identification of cytokines, chemokines,

    growth factors, and various other proliferation factors in

    LCH, avenues for employing new therapeutic agents

    become available.17,82,83,87-94 One such agent is 2-CDA

    Table XI. Treatment protocols and late effects inLangerhans cell histiocytosis*

    Treatment protocols by The Histiocyte Society

    LCH-I pediatric clinical trial: multisystem disease

    Vinblastine and intravenous steroid or

    Etoposide (possible risk of second malignancy)

    and intravenous steroid

    Prognostic factors in pediatric LCH-II clinical trialTherapeutic response at 6 weeks in high-risk patients

    Good response: 94% survival

    Poor response: 34% survival

    LCH-II pediatric clinical trial: multisystem disease

    for high-risk sites

    Vinblastine, oral prednisone, mercaptopurine or

    Vinblastine,oral prednisone,mercaptopurine, etoposide

    Prognostic factors in pediatric LCH-II clinical trial

    Therapeutic response at 6 weeks in high-risk patients

    Good response: 88% survival

    Poor response: 17% survival

    Age: No effect on survival

    LCH-III pediatric randomized clinical trial

    Multisystem disease with high-risk sites

    (lung, liver, bone marrow, spleen)

    A: Vinblastine and prednisone for 12 months

    B: Vinblastine, prednisone, and methotrexate

    (1st 6 weeks),

    Followed by maintenance vinblastine and

    methotrexate, daily oral mercaptopurine and

    oral methotrexate weekly for 12 months

    Multisystem disease with low-risk sites

    (skin, bone, lymph nodes, pituitary)

    Vinblastine and prednisone (6 or 12 months)

    Single system multifocal disease

    Localized special sites (mastoid, orbit, temporal bones)

    LCH-A-I adult clinical trial

    Single system disease

    CNS disease: vinblastine and prednisone

    Isolated pulmonary disease: vinblastine and prednisone

    Multisystem disease: vinblastine and prednisone

    Current treatment reported for those not on The Histiocyte

    Association LCH protocols

    Unifocal osseous lesion

    Surgical curettage or excision

    Radiation therapy

    Single agent chemotherapy

    Combination of above

    Multisystem, refractory, relapsing or progressive disease

    Chemotherapy with or without radiation therapy

    Comparison of bone disease recurrence

    Multiagent chemotherapyBone-only disease: 18% recurrence

    (unifocal or multifocal)

    Single-agent chemotherapy, radiotherapy

    or observation only

    Bone-only disease: 50%-80% recurrence

    Late effects among long-term pediatric survivors, %

    At least 1 permanent consequence

    All groups 52

    Multisystem disease 71

    Single system disease 24

    Diabetes insipidus 24

    Table XI. Continued

    Orthopedic abnormalities 20

    Hearing loss 13

    Neurologic consequences 11

    Growth retardation 9

    Ophthalmic problems 8

    Tooth loss 7

    Pulmonary problems 4Skin problems 2

    Innovative therapeutic agents

    2-CDA (2-chloro-29-deoxyadenosine, cladribine)

    Deoxycoformycin

    Thalidomide

    Alemtuzumab (anti-CD52 antibody)

    Retinoic acid

    Interleukin-2

    Interferon-alpha

    Cyclosporin A

    Tacrolimus (FK506)

    Monoclonal antibody against CD1a and CD207

    (langerin)

    Intralesional injection of steroids or chemotherapeutic

    agent

    Phototherapy (ultraviolet light treatment for skin disease)

    Bone marrow transplantation (allogeneic or autologous

    stem cell)

    LCH, Langerhans cell histiocytosis; CNS, central nervous system.

    *Compiled from references15, 17, 82-95.

    OOOOE

    S62 Hicks and Flaitz August 2005

    http://-/?-http://-/?-
  • 7/21/2019 LCH Sunil Sir

    22/25

    (2-chloro-29-deoxyadenosine; cladribine).87 This agent

    has previously shown good results with lymphoblastic

    leukemia, hairy-cell leukemia, chronic lymphocytic leu-

    kemia, and Waldenstroms macroglobulinemia. Initial

    studies in children with refractory or progressive multi-

    focal bone disease or multiorgan disease involvement

    have shown promise. All patients had a clinical responsewith 70% requiring no additional therapy after a median

    of 50 months follow-up. The other 30% required addi-

    tional therapy to become disease-free. Because 2-CDA

    is a potent immunosuppressive agent, toxicity was

    limited to manageable myelosuppression. Deoxycofor-

    mycin is a therapeutic agent related to 2-CDA, which

    has been used to a limited extent in other neoplastic

    processes, and may be worth further investigation in

    LCH.

    Thalidomide has been used as a therapeutic agent

    with cutaneous LCH with considerable success.88,89 In

    13 adult patients, good responses were noted with

    resolution of ulcers located on the skin and inguinal,

    genital, and perianal areas, with replacement by scar

    tissue. The lesions regressed within 1 to 3 months, butrecurred if oral thalidomide treatment was discontinued.

    None of the patients reported peripheral neuropathy.

    The teratogenic side effect of this drug is well known,

    and birth control is mandated in females of child-

    bearing age. Thalidomide is an inhibitor of TNF-alpha

    and IL-6. Both of these cellular factors participate in

    LCH disease. Of interest, another TNF-alpha inhibitor,

    etanercept, has shown similar effects in a single case

    study.

    A monoclonal antibody against CD52 (alemtuzumab)has recently been developed, and is effective indeplet-

    ing lymphocytes and dendritic cells in vivo.90 Quite

    exciting is the finding that CD52 is expressed on

    Langerhans cell histiocytes, but not on normal

    Langerhans cells within the epidermis. This finding is

    encouraging and provides potential targeted therapy for

    patients with refractory high-risk disease resistant to

    current therapy. Alemtuzumab has been well tolerated

    in clinical trials with refractory chronic lymphocytic

    leukemia patients.

    Retinoic acid has the ability to induce differentiation

    in several different cell types, including leukemic cells,neuroblastoma cells, and Langerhans cell histiocytes

    in vitro.91,92 In patients with LCH, retinoic acid hasinduced remission in some cases and stabilized disease

    progression in others. Additional coordinated studies

    are necessary to determine the therapeutic potential of

    this agent in LCH.

    Several other therapeutic agents may induce immune

    modulation (IL-2, IFN-alpha) or suppress the immune

    system (cyclosporine A, tacrolimus).73,79-81-83,93,94

    Monoclonal antibodies directed against CD1a or per-

    haps even CD207 (langerin) may be employed as

    treatment in the near future. Individual case reports have

    shown regression of LCH lesions with intralesional

    steroid and chemotherapeutic agent administration.17,95

    The role of bone marrow transplantation and stem cell

    reconstitution are still under investigation as treatment

    adjuncts for refractory high-risk patients.

    17,82,83

    SUMMARYWhen the sentinel case report of eosinophilic gran-

    uloma of the jaw with anal involvement was published

    almost 6 decades ago, this entity had only recently been

    included in the new category of Histiocytosis X.3 The

    cell of origin was not known, however the transitive

    phases between eosinophilic granuloma of the bone and

    Letterer-Siwe disease was noted by the author due to

    the histologic picture of both lesions showing eosin-

    ophilic infiltration in the tissue3 (p. 256). As noted in the

    current review, Langerhans cell histiocytes have been

    determined to be the prime component of this disease. It

    is also obvious that this cell is an immature dendritic cell

    and not a histiocyte at all.

    No doubt the extended period from presentation to

    diagnosis with Schroffs case3 was related to the rarity

    of such a lesion and lack of familiarity with this entity

    by pathologists some 60 years ago. Only about 65 cases

    were described in the worlds literature at that time.3

    Although a rare orphan disease, LCH is readily

    identified now by general and oral and maxillofacial

    pathologists, as well as by residents in-training. The

    clinical and radiographic features in the affected

    gentleman are identical to those seen in patients withLCH today. While more sophisticated diagnostic imag-

    ing techniques are available, current dental practitioners

    use the same radiographic methods used more than 60

    years ago to evaluate patients for jaw lesions. Many

    sophisticated diagnostic tools are available to modern

    day practitioners, such as computed tomography (CT),

    magnetic resonance imaging (MRI), nuclear bone scans,

    and positron emission tomography (PET). These may be

    used to define the jaw lesions or for detection of other

    organ and bone involvement.

    Confirmation of the diagnosis of LCH has changed

    considerably since the early 1940s. Electron micros-copy became available as a diagnostic tool in the early

    1950s and was widely adopted in the late 1960s. A

    lesion-defining ultrastructural feature for Langerhans

    cell histiocytes was discovered in the early 1960sthe

    Birbeck granule. The identification of this structure in

    both normal Langerhans cells in the epidermis and in

    histiocytes within lesional tissue provided the evi-

    dence for the cell of origin for LCH. At the time of

    Schroffs case report in 1948,3 immunocytochemical

    techniques with tissue sections were not yet available.

    OOOOE

    Volume 100, Number 2 Hicks and Flaitz S63

  • 7/21/2019 LCH Sunil Sir

    23/25

    By the late 1970s, several immunocytochemical anti-

    bodies had been developed for diagnostic purposes, but

    required that lesional tissue be frozen at 708C.