National Cancer Institute
U.S. National Institutes of Health | www.cancer.gov

NCI Home
Cancer Topics
Clinical Trials
Cancer Statistics
Research & Funding
News
About NCI
Langerhans Cell Histiocytosis (PDQ®)
Health Professional Version   Last Modified: 04/28/2009



Purpose of This PDQ Summary






General Information






Childhood Langerhans Cell Histiocytosis






Histopathologic, Immunologic, and Cytogenetic Characteristics of Langerhans Cell Histiocytosis






Presentation of Langerhans Cell Histiocytosis in Children






Diagnostic Evaluation of Childhood Langerhans Cell Histiocytosis






Follow-up Considerations in Childhood Langerhans Cell Histiocytosis






Treatment of Childhood Langerhans Cell Histiocytosis






Treatment of Recurrent, Refractory or Progressive Childhood Langerhans Cell Histiocytosis






Late Disease and Treatment Effects of Childhood Langerhans Cell Histiocytosis






Adult Langerhans Cell Histiocytosis






Treatment of Adult Langerhans Cell Histiocytosis






Get More Information From NCI






Changes to the Summary (04/28/2009)






More Information



Page Options
Print This Page
Print Entire Document
View Entire Document
E-Mail This Document
Quick Links
Director's Corner

Dictionary of Cancer Terms

NCI Drug Dictionary

Funding Opportunities

NCI Publications

Advisory Boards and Groups

Science Serving People

Español
Quit Smoking Today
NCI Highlights
The Nation's Investment in Cancer Research FY 2010

Report to Nation Finds Declines in Cancer Incidence, Death Rates

High Dose Chemotherapy Prolongs Survival for Leukemia

Prostate Cancer Study Shows No Benefit for Selenium, Vitamin E
Histopathologic, Immunologic, and Cytogenetic Characteristics of Langerhans Cell Histiocytosis

Cell of Origin and Biologic Correlates
Immunologic Abnormalities
Etiology
Chromosomal Studies
Cytokine Analysis by Immunohistochemical Staining
Gene Expression by Microarray Analysis
Cytokine Levels and Association with Diagnosis, Prognosis or Response to Therapy
Human Leukocyte Antigen Type and Association with Langerhans Cell Histiocytosis



Cell of Origin and Biologic Correlates

The Langerhans cell (LC) originates from bone marrow stem cells as an immature dendritic cell (DC) which can then develop into the LC under the influence of several cytokines, including granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor alpha (TNF alpha), and others.[1] These immature cells are found in the skin, lymph nodes, spleen, bone marrow, and lungs. LCs differ from other histiocytes in that they are CD1a-positive and CD207 (langerin)-positive.[2] This cell characteristic along with the classic histology (see below) is used to identify LCs in lesions where they also maintain an immature dendritic cell phenotype (see below).[3] Suspected Langerhans cell histiocytosis (LCH) lesions also contain lymphocytes, macrophages, neutrophils, eosinophils, fibroblasts and sometimes multinucleated giant cells. It has been proposed that the LC is the cellular pathologic culprit for LCH lesions, however, definitive evidence of this has yet to be obtained.[2]

In the brain, three types of histopathologic findings have been described in LCH:

Granulomas in meninges or choroid plexus with CD1a+ LC and predominantly CD8+ lymphocytes.


Granulomas in connective tissue spaces with CD1a+ LC and predominantly CD8+lymphocytes causing an inflammatory response and neuronal loss.


Predominantly CD8+ lymphocyte infiltration with tissue degeneration, microglial activation and gliosis.[4]


Immunologic Abnormalities

Normally, the LC is a primary presenter of antigen to naïve T lymphocytes. However, in LCH, the LC does not efficiently stimulate primary T lymphocyte responses.[5] Antibody staining for the DC markers, CD80, CD86, and class II antigens, has been used to show that in LCH, the abnormal cells are immature DCs that present antigen poorly and are proliferating at a low rate.[3,6] Transforming growth factor-beta (TGF-beta) as well as interleukin (IL)-10 are possibly responsible for preventing LC maturation in LCH.[3] The expansion of regulatory T cells in LCH patients has been reported.[6] The population of CD4+ CD25 high FoxP3high cells was reported to comprise 20% of T cells and appeared to be in contact with LC in LCH lesions. These T cells were present in higher numbers in the peripheral blood of LCH patients than in controls and returned to a normal level when patients were in remission.

Etiology

The etiology of LCH is unknown. Efforts to define a viral cause have not been successful.[7,8]

Chromosomal Studies

Studies showing clonality in LCH have been published since 1997 using polymorphisms of methylation-specific restriction enzyme sites on the X-chromosome regions coding for the human androgen receptor, DXS255, PGK, and HPRT.[9,10] Biopsies of lesions with single system or multisystem disease were found to have a proliferation of LCs from a single clone. Pulmonary LCH in adults is usually nonclonal.[11] Cytogenetic abnormalities in LCH have rarely been reported. One study described an abnormal clone (t7;12)(q11.2;p13) from a vertebral lesion of one patient.[12] This study also reported nonclonal karyotypic abnormalities in three patients. An increase in chromosomal breakage was also noted.

Three studies have used comparative genomic hybridization (CGH) to analyze bone and pulmonary LCH.[13-15] Analysis of seven bone lesions by CGH and loss of heterozygosity (LOH) has provided further evidence to suggest that chromosomal aberrations may be an intrinsic problem in LCH.[13] One study evaluated 14 cases of pulmonary LCH for LOH [14] and found LOH of 1p, 1q, 3p, 5p, 17p and 22q. Allelic loss of one or more tumor suppressor genes was identified in 19 of 24 specimens. Researchers at the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins demonstrated that significantly shortened telomeres in the LCH lesional LCs compared with LCs in inflammatory disorders such as dermatopathic lymphadenitis.[16] An Italian group demonstrated that high-risk LCH patients had DNA polymorphisms of two cytokine genes (IL-4 and interferon gamma) which were associated with high-expresser phenotypes.[17]

Cytokine Analysis by Immunohistochemical Staining

Several cytokines have apparent higher expression levels in LCH lesions than in normal tissue. Among the cytokines expressed at high levels are GM-CSF, interferon gamma, IL-1, sIL-2r, IL-10, and the TNF receptor family member, RANK.[6,18] Lesional T cells stain with antibodies to TNF alpha, various interleukins, CD40L, and sCD154.[18,19] Prominent expression of angiotensin-converting enzyme, TGF-beta, and IL-11 in LCH biopsies have been reported.[20] Increased expression of the chemokine receptor, CCR6, has been observed in LCH lesions and may contribute to the accumulation of LCs in skin. Increased CCR7 expression may cause the LC to home to lymphoid tissue.[21] These data are somewhat anomalous because it is expected that CCR6 would be down-regulated in the activated LCs in these lesions. However, prominent staining of CCR6 and its ligand CCL20/M1P-3alpha has been found in other studies.[22] Further evidence for the altered biology of LCs in LCH is the increased expression of the antiapoptotic protein, Bcl-2, as well as several other genes (e.g., Ki-67, TGF-beta receptors 1 and 2, MDM2, p53, p21, p16, and RB).[23]

Gene Expression by Microarray Analysis

Two papers have been published on analysis of gene expression in LCH by gene array techniques.[24,25] In the first report, a laser-capture microdissection was used to purify LCs from frozen biopsy specimens or normal LCs from the skin.[24] The level of cytokine and growth factor gene expression in the control LCs versus those from patient biopsies showed a striking similarity in expression of RNA of the TNF family of genes and several interleukins. Only macrophage colony-stimulating factor (M-CSF), TGF-beta receptor and IL-1 alpha transcripts were expressed at higher levels in the LCs from LCH patients. In the second report, a very different approach was used, generating LCs from CD34 stem cells in vitro followed by a serial analysis of gene expression library to identify highly expressed genes in these LCs.[25] Several of the highly expressed genes were then chosen to test for RNA levels from whole LCH biopsy specimens. High expression of FSCN1, GSN MMP12, CCL22, CD1a and CD207 was commonly observed.

Cytokine Levels and Association with Diagnosis, Prognosis or Response to Therapy

Several groups have measured cytokines or other markers of immune activation in the plasma or serum of patients with LCH to determine whether these levels could be of diagnostic or prognostic importance or whether the levels could be helpful for evaluating response to therapy. One group reported consistently higher levels of IL-2 receptor in LCH biopsy specimens,[26] however, they found no increased expression of IL-1beta, TNF-alpha, and IL-2, which were all within normal limits. Another group used a similar assay technique and identified increased levels of the IL-1 receptor agonist and TNF-alpha in plasma from LCH patients.[27] Vascular endothelial growth factor has also been identified by immunohistochemistry in five of five patients with multisystem LCH and two of five patients with single system LCH.[28] A third group found markedly elevated levels of FLT3-ligand and M-CSF in the serum of LCH patients with good correlation to the extent of disease as well as response to treatment and increased circulating immature myeloid dendritic cells.[29] It should be noted that this latter finding was not confirmed by a later study; however, there was a trend toward higher myeloid DC levels in patients.[6] Elevated levels of another regulator of the immune system and bone metabolism, osteoprotegerin (OPG), are present in the plasma of patients with active LCH.[30,31] OPG levels reported in this study were highest in patients with multisystem disease and decreased with response to therapy, similar to the findings with FLT-3 ligand and M-CSF.

Human Leukocyte Antigen Type and Association with Langerhans Cell Histiocytosis

Specific associations of LCH with distinct human lymphocyte antigen (HLA) types and extent of disease have been published. In a study of 84 Nordic patients, those with only skin or bone involvement more frequently had HLA-DRB1*03 type than those with multisystem disease.[32] In 29 patients and 37 family members in the United States, the Cw7 and DR4 types were significantly more prevalent in Caucasians with single bone lesions.[33]

References

  1. Laman JD, Leenen PJ, Annels NE, et al.: Langerhans-cell histiocytosis 'insight into DC biology'. Trends Immunol 24 (4): 190-6, 2003.  [PUBMED Abstract]

  2. Weitzman S, Egeler R M, eds.: Histiocytic Disorders of Children and Adults. Cambridge, United Kingdom: Cambridge University Press, 2005. 

  3. Geissmann F, Lepelletier Y, Fraitag S, et al.: Differentiation of Langerhans cells in Langerhans cell histiocytosis. Blood 97 (5): 1241-8, 2001.  [PUBMED Abstract]

  4. Grois N, Prayer D, Prosch H, et al.: Neuropathology of CNS disease in Langerhans cell histiocytosis. Brain 128 (Pt 4): 829-38, 2005.  [PUBMED Abstract]

  5. Yu RC, Morris JF, Pritchard J, et al.: Defective alloantigen-presenting capacity of 'Langerhans cell histiocytosis cells'. Arch Dis Child 67 (11): 1370-2, 1992.  [PUBMED Abstract]

  6. Senechal B, Elain G, Jeziorski E, et al.: Expansion of regulatory T cells in patients with Langerhans cell histiocytosis. PLoS Med 4 (8): e253, 2007.  [PUBMED Abstract]

  7. McClain K, Jin H, Gresik V, et al.: Langerhans cell histiocytosis: lack of a viral etiology. Am J Hematol 47 (1): 16-20, 1994.  [PUBMED Abstract]

  8. Glotzbecker MP, Dormans JP, Pawel BR, et al.: Langerhans cell histiocytosis and human herpes virus 6 (HHV-6), an analysis by real-time polymerase chain reaction. J Orthop Res 24 (3): 313-20, 2006.  [PUBMED Abstract]

  9. Willman CL, Busque L, Griffith BB, et al.: Langerhans'-cell histiocytosis (histiocytosis X)--a clonal proliferative disease. N Engl J Med 331 (3): 154-60, 1994.  [PUBMED Abstract]

  10. Yu RC, Chu C, Buluwela L, et al.: Clonal proliferation of Langerhans cells in Langerhans cell histiocytosis. Lancet 343 (8900): 767-8, 1994.  [PUBMED Abstract]

  11. Yousem SA, Colby TV, Chen YY, et al.: Pulmonary Langerhans' cell histiocytosis: molecular analysis of clonality. Am J Surg Pathol 25 (5): 630-6, 2001.  [PUBMED Abstract]

  12. Betts DR, Leibundgut KE, Feldges A, et al.: Cytogenetic abnormalities in Langerhans cell histiocytosis. Br J Cancer 77 (4): 552-5, 1998.  [PUBMED Abstract]

  13. Murakami I, Gogusev J, Fournet JC, et al.: Detection of molecular cytogenetic aberrations in langerhans cell histiocytosis of bone. Hum Pathol 33 (5): 555-60, 2002.  [PUBMED Abstract]

  14. Dacic S, Trusky C, Bakker A, et al.: Genotypic analysis of pulmonary Langerhans cell histiocytosis. Hum Pathol 34 (12): 1345-9, 2003.  [PUBMED Abstract]

  15. Chikwava KR, Hunt JL, Mantha GS, et al.: Analysis of loss of heterozygosity in single-system and multisystem Langerhans' cell histiocytosis. Pediatr Dev Pathol 10 (1): 18-24, 2007 Jan-Feb.  [PUBMED Abstract]

  16. Bechan GI, Meeker AK, De Marzo AM, et al.: Telomere length shortening in Langerhans cell histiocytosis. Br J Haematol 140 (4): 420-8, 2008.  [PUBMED Abstract]

  17. De Filippi P, Badulli C, Cuccia M, et al.: Specific polymorphisms of cytokine genes are associated with different risks to develop single-system or multi-system childhood Langerhans cell histiocytosis. Br J Haematol 132 (6): 784-7, 2006.  [PUBMED Abstract]

  18. Egeler RM, Favara BE, van Meurs M, et al.: Differential In situ cytokine profiles of Langerhans-like cells and T cells in Langerhans cell histiocytosis: abundant expression of cytokines relevant to disease and treatment. Blood 94 (12): 4195-201, 1999.  [PUBMED Abstract]

  19. Egeler RM, Favara BE, Laman JD, et al.: Abundant expression of CD40 and CD40-ligand (CD154) in paediatric Langerhans cell histiocytosis lesions. Eur J Cancer 36 (16): 2105-10, 2000.  [PUBMED Abstract]

  20. Brown RE: Angiotensin-converting enzyme, transforming growth factor beta(1), and interleukin 11 in the osteolytic lesions of Langerhans cell histiocytosis. Arch Pathol Lab Med 124 (9): 1287-90, 2000.  [PUBMED Abstract]

  21. Fleming MD, Pinkus JL, Fournier MV, et al.: Coincident expression of the chemokine receptors CCR6 and CCR7 by pathologic Langerhans cells in Langerhans cell histiocytosis. Blood 101 (7): 2473-5, 2003.  [PUBMED Abstract]

  22. Annels NE, Da Costa CE, Prins FA, et al.: Aberrant chemokine receptor expression and chemokine production by Langerhans cells underlies the pathogenesis of Langerhans cell histiocytosis. J Exp Med 197 (10): 1385-90, 2003.  [PUBMED Abstract]

  23. Schouten B, Egeler RM, Leenen PJ, et al.: Expression of cell cycle-related gene products in Langerhans cell histiocytosis. J Pediatr Hematol Oncol 24 (9): 727-32, 2002.  [PUBMED Abstract]

  24. McClain KL, Cai YH, Hicks J, et al.: Expression profiling using human tissues in combination with RNA amplification and microarray analysis: assessment of Langerhans cell histiocytosis. Amino Acids 28 (3): 279-90, 2005.  [PUBMED Abstract]

  25. Rust R, Kluiver J, Visser L, et al.: Gene expression analysis of dendritic/Langerhans cells and Langerhans cell histiocytosis. J Pathol 209 (4): 474-83, 2006.  [PUBMED Abstract]

  26. Schultz C, Klouche M, Friedrichsdorf S, et al.: Langerhans cell histiocytosis in children: does soluble interleukin-2-receptor correlate with both disease extent and activity? Med Pediatr Oncol 31 (2): 61-5, 1998.  [PUBMED Abstract]

  27. Rosso DA, Ripoli MF, Roy A, et al.: Serum levels of interleukin-1 receptor antagonist and tumor necrosis factor-alpha are elevated in children with Langerhans cell histiocytosis. J Pediatr Hematol Oncol 25 (6): 480-3, 2003.  [PUBMED Abstract]

  28. Pavlakovic H, Von Schütz V, Rössler J, et al.: Quantification of angiogenesis stimulators in children with solid malignancies. Int J Cancer 92 (5): 756-60, 2001.  [PUBMED Abstract]

  29. Rolland A, Guyon L, Gill M, et al.: Increased blood myeloid dendritic cells and dendritic cell-poietins in Langerhans cell histiocytosis. J Immunol 174 (5): 3067-71, 2005.  [PUBMED Abstract]

  30. Ishii R, Morimoto A, Ikushima S, et al.: High serum values of soluble CD154, IL-2 receptor, RANKL and osteoprotegerin in Langerhans cell histiocytosis. Pediatr Blood Cancer 47 (2): 194-9, 2006.  [PUBMED Abstract]

  31. Rosso DA, Karis J, Braier JL, et al.: Elevated serum levels of the decoy receptor osteoprotegerin in children with langerhans cell histiocytosis. Pediatr Res 59 (2): 281-6, 2006.  [PUBMED Abstract]

  32. Bernstrand C, Carstensen H, Jakobsen B, et al.: Immunogenetic heterogeneity in single-system and multisystem langerhans cell histiocytosis. Pediatr Res 54 (1): 30-6, 2003.  [PUBMED Abstract]

  33. McClain KL, Laud P, Wu WS, et al.: Langerhans cell histiocytosis patients have HLA Cw7 and DR4 types associated with specific clinical presentations and no increased frequency in polymorphisms of the tumor necrosis factor alpha promoter. Med Pediatr Oncol 41 (6): 502-7, 2003.  [PUBMED Abstract]

Back to Top

< Previous Section  |  Next Section >


A Service of the National Cancer Institute
Department of Health and Human Services National Institutes of Health USA.gov