Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-pjpqr Total loading time: 0 Render date: 2024-06-28T20:45:38.242Z Has data issue: false hasContentIssue false

Chapter 28 - Histiocytic Inflammatory Neoplasms/Lesions

from Section VI - Histiocytic Disorders and Neoplasms

Published online by Cambridge University Press:  25 January 2024

Xiayuan Liang
Affiliation:
Children’s Hospital of Colorado
Bradford Siegele
Affiliation:
Children’s Hospital of Colorado
Jennifer Picarsic
Affiliation:
Cincinnati Childrens Hospital Medicine Center
Get access

Summary

Langerhans cell histiocytosis (LCH), juvenile xanthogranuloma (JXG), and Rosai-Dorfman-Destombes disease (RDD) can each manifest as a focal lesion, as multiple lesions, or as a widespread systemic disorder with organ involvement. Erdheim-Chester disease (ECD) is a rare systemic disease process in children, with more frequent adult presentations. New distinct emerging entities covered include ALK-positive histiocytosis and post-leukemia/lymphoma histiocytic lesions. These histiocytic lesions are now best classified as myeloid-derived inflammatory neoplastic disorders composed of clonal dendritic- or macrophage-/monocyte-derived cells that infiltrate tissues and are driven by recurrent kinase-activating alterations, most often in the mitogen-activated protein kinase (MAPK), PI3K-AKT, and receptor tyrosine kinase (RTK) signaling pathways, which have all had a long history of being associated with human neoplasia, with ERK overexpression noted in many of these neoplasms (Fig. 28.1; Table 28.1) (1–3). Furthermore, these oncological signaling cascades are critical to the intranuclear regulation of transcription factors that serve as key factors influencing cellular proliferation, survival, and differentiation (4).

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2024

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Berres, ML, Lim, KP, Peters, T, Price, J, Takizawa, H, Salmon, H, et al. BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups. J Exp Med. 2014;211(4):669–83.Google Scholar
Durham, BH, Roos-Weil, D, Baillou, C, Cohen-Aubart, F, Yoshimi, A, Miyara, M, et al. Functional evidence for derivation of systemic histiocytic neoplasms from hematopoietic stem/progenitor cells. Blood. 2017;130(2):176–80.Google Scholar
Abla, O, Jacobsen, E, Picarsic, J, Krenova, Z, Jaffe, R, Emile, JF, et al. Consensus recommendations for the diagnosis and clinical management of Rosai-Dorfman-Destombes disease. Blood. 2018;131(26):2877–90.Google Scholar
Wellbrock, C, Karasarides, M, Marais, R. The RAF proteins take centre stage. Nat Rev Mol Cell Biol. 2004;5(11):875–85.Google Scholar
Emile, JF, Abla, O, Fraitag, S, Horne, A, Haroche, J, Donadieu, J, et al. Revised classification of histiocytoses and neoplasms of the macrophage-dendritic cell lineages. Blood. 2016;127(22):2672–81.Google Scholar
Lim, KPH, Milne, P, Poidinger, M, Duan, K, Lin, H, McGovern, N, et al. Circulating CD1c+ myeloid dendritic cells are potential precursors to LCH lesion CD1a+CD207+ cells. Blood Adv. 2020;4(1):8799.Google Scholar
Goyal, G, Heaney, ML, Collin, M, Cohen-Aubart, F, Vaglio, A, Durham, BH, et al. Erdheim-Chester disease: consensus recommendations for evaluation, diagnosis, and treatment in the molecular era. Blood. 2020;135(22):1929–45.Google Scholar
Guyot-Goubin, A, Donadieu, J, Barkaoui, M, Bellec, S, Thomas, C, Clavel, J. Descriptive epidemiology of childhood Langerhans cell histiocytosis in France, 2000–2004. Pediatr Blood Cancer. 2008;51(1):71–5.Google Scholar
Krooks, J, Minkov, M, Weatherall, AG. Langerhans cell histiocytosis in children: history, classification, pathobiology, clinical manifestations, and prognosis. J Am Acad Dermatol. 2018;78(6):1035–44.Google Scholar
Krooks, J, Minkov, M, Weatherall, AG. Langerhans cell histiocytosis in children: diagnosis, differential diagnosis, treatment, sequelae, and standardized follow-up. J Am Acad Dermatol. 2018;78(6):1047–56.Google Scholar
Heritier, S, Emile, JF, Barkaoui, MA, Thomas, C, Fraitag, S, Boudjemaa, S, et al. BRAF mutation correlates with high-risk Langerhans cell histiocytosis and increased resistance to first-line therapy. J Clin Oncol. 2016;34(25):3023–30.Google Scholar
Lee, LH, Krupski, C, Clark, J, Wunderlich, M, Lorsbach, RB, Grimley, MS, et al. High-risk LCH in infants is serially transplantable in a xenograft model but responds durably to targeted therapy. Blood Adv. 2020;4(4):717–27.Google Scholar
Gadner, H, Minkov, M, Grois, N, Potschger, U, Thiem, E, Arico, M, et al. Therapy prolongation improves outcome in multisystem Langerhans cell histiocytosis. Blood. 2013;121(25):5006–14.Google Scholar
Haupt, R, Minkov, M, Astigarraga, I, Schafer, E, Nanduri, V, Jubran, R, et al. Langerhans cell histiocytosis (LCH): guidelines for diagnosis, clinical work-up, and treatment for patients till the age of 18 years. Pediatr Blood Cancer. 2013;60(2):175–84.Google Scholar
Allen, CE, Ladisch, S, McClain, KL. How I treat Langerhans cell histiocytosis. Blood. 2015;126(1):2635.Google Scholar
Grois, N, Potschger, U, Prosch, H, Minkov, M, Arico, M, Braier, J, et al. Risk factors for diabetes insipidus in Langerhans cell histiocytosis. Pediatr Blood Cancer. 2006;46(2):228–33.Google Scholar
Geissmann, F, Lepelletier, Y, Fraitag, S, Valladeau, J, Bodemer, C, Debre, M, et al. Differentiation of Langerhans cells in Langerhans cell histiocytosis. Blood. 2001;97(5):1241–8.Google Scholar
Allen, CE, Li, L, Peters, TL, Leung, HC, Yu, A, Man, TK, et al. Cell-specific gene expression in Langerhans cell histiocytosis lesions reveals a distinct profile compared with epidermal Langerhans cells. J Immunol. 2010;184(8):4557–67.Google Scholar
Rollins, BJ. Genomic alterations in Langerhans cell histiocytosis. Hematol Oncol Clin North Am. 2015;29(5):839–51.Google Scholar
Allen, CE, Merad, M, McClain, KL. Langerhans-cell histiocytosis. N Engl J Med. 2018;379(9):856–68.Google Scholar
Xiao, Y, van Halteren, AG, Lei, X, Borst, J, Steenwijk, EC, de Wit, T, et al. Bone marrow-derived myeloid progenitors as driver mutation carriers in high-and low-risk Langerhans cell histiocytosis. Blood. 2020;136(19):2188–99.Google Scholar
Picarsic, J, Jaffe, R. Nosology and pathology of Langerhans cell histiocytosis. Hematol Oncol Clin North Am. 2015;29(5):799823.Google Scholar
Abdallah, M, Généreau, T, Donadieu, J, Emile, JF, Chazouillères, O, Gaujoux-Viala, C, et al. Langerhans’ cell histiocytosis of the liver in adults. Clin Res Hepatol Gastroenterol. 2011;35(6–7):475–81.Google Scholar
Kaplan, KJ, Goodman, ZD, Ishak, KG. Liver involvement in Langerhans’ cell histiocytosis: a study of nine cases. Mod Pathol. 1999;12(4):370–8.Google Scholar
Kambouchner, M, Emile, JF, Copin, MC, Coulomb-Lherminé, A, Sabourin, JC, Della Valle, V, et al. Childhood pulmonary Langerhans cell histiocytosis: a comprehensive clinical-histopathological and BRAF(V600E) mutation study from the French national cohort. Hum Pathol. 2019;89:5161.Google Scholar
Favara, BE, Jaffe, R, Egeler, RM. Macrophage activation and hemophagocytic syndrome in Langerhans cell histiocytosis: report of 30 cases. Pediatr Dev Pathol. 2002;5(2):130–40.Google Scholar
Chellapandian, D, Hines, MR, Zhang, R, Jeng, M, van den Bos, C, Santa-María López, V, et al. A multicenter study of patients with multisystem Langerhans cell histiocytosis who develop secondary hemophagocytic lymphohistiocytosis. Cancer. 2019;125(6):963–71.Google Scholar
Shanmugam, V, Craig, JW, Hornick, JL, Morgan, EA, Pinkus, GS, Pozdnyakova, O. Cyclin D1 is expressed in neoplastic cells of Langerhans cell histiocytosis but not reactive Langerhans cell proliferations. Am J Surg Pathol. 2017;41(10):1390–6.Google Scholar
Willman, CL. Detection of clonal histiocytes in Langerhans cell histiocytosis: biology and clinical significance. Br J Cancer Suppl. 1994;23:S29S33.Google Scholar
Chetritt, J, Paradis, V, Dargere, D, Adle-Biassette, H, Maurage, CA, Mussini, JM, et al. Chester-Erdheim disease: a neoplastic disorder. Human Pathology. 1999;30(9):1093–6.Google Scholar
Badalian-Very, G, Vergilio, JA, Degar, BA, MacConaill, LE, Brandner, B, Calicchio, ML, et al. Recurrent BRAF mutations in Langerhans cell histiocytosis. Blood. 2010;116(11):1919–23.Google Scholar
Durham, BH. Molecular characterization of the histiocytoses: neoplasia of dendritic cells and macrophages. Seminars in Cell & Developmental Biology. 2019;86:6276.Google Scholar
Durham, BH, Lopez Rodrigo, E, Picarsic, J, Abramson, D, Rotemberg, V, De Munck, S, et al. Activating mutations in CSF1R and additional receptor tyrosine kinases in histiocytic neoplasms. Nat Med. 2019;25(12):1839–42.Google Scholar
Hogstad, B, Berres, ML, Chakraborty, R, Tang, J, Bigenwald, C, Serasinghe, M, et al. RAF/MEK/extracellular signal-related kinase pathway suppresses dendritic cell migration and traps dendritic cells in Langerhans cell histiocytosis lesions. J Exp Med. 2018;215(1):319–36.Google Scholar
Bigenwald, C, Le Berichel, J, Wilk, CM, Chakraborty, R, Chen, ST, Tabachnikova, A, et al. BRAF(V600E)-induced senescence drives Langerhans cell histiocytosis pathophysiology. Nat Med. 2021;27(5):851–61.Google Scholar
Bhattacharjee, P, Glusac, EJ. Langerhans cell hyperplasia in scabies: a mimic of Langerhans cell histiocytosis. J Cutan Pathol. 2007;34(9):716–20.Google Scholar
Picarsic, J, Jaffe, R. Pathology of histiocytic disorders and neoplasms and related disorders. In: Abla, O, Janka, G, eds. Histiocytic disorders. Cham, Switzerland: Springer International; 2018: 350.Google Scholar
Favara, BE, Steele, A. Langerhans cell histiocytosis of lymph nodes: a morphological assessment of 43 biopsies. Pediatr Pathol Lab Med. 1997;17(5):769–87.Google Scholar
Edelweiss, M, Medeiros, LJ, Suster, S, Moran, CA. Lymph node involvement by Langerhans cell histiocytosis: a clinicopathologic and immunohistochemical study of 20 cases. Hum Pathol. 2007;38(10):1463–9.Google Scholar
Garces, S, Yin, CC, Miranda, RN, Patel, KP, Li, S, Xu, J, et al. Clinical, histopathologic, and immunoarchitectural features of dermatopathic lymphadenopathy: an update. Mod Pathol. 2020;33(6):1104–21.Google Scholar
Geissmann, F, Dieu-Nosjean, MC, Dezutter, C, Valladeau, J, Kayal, S, Leborgne, M, et al. Accumulation of immature Langerhans cells in human lymph nodes draining chronically inflamed skin. J Exp Med. 2002;196(4):417–30.Google Scholar
Chikwava, K, Jaffe, R. Langerin (CD207) staining in normal pediatric tissues, reactive lymph nodes, and childhood histiocytic disorders. Pediatr Dev Pathol. 2004;7(6):607–14.Google Scholar
Selove, W, Picarsic, J, Swerdlow, SH. Langerin staining identifies most littoral cell angiomas but not most other splenic angiomatous lesions. Hum Pathol. 2019;83:43–9.Google Scholar
Titgemeyer, C, Grois, N, Minkov, M, Flucher-Wolfram, B, Gatterer-Menz, I, Gadner, H. Pattern and course of single-system disease in Langerhans cell histiocytosis data from the DAL-HX 83- and 90-study. Med Pediatr Oncol. 2001;37(2):108–14.Google Scholar
Zelger, BW, Sidoroff, A, Orchard, G, Cerio, R. Non-Langerhans cell histiocytoses. A new unifying concept. Am J Dermatopathol. 1996;18(5):490504.Google Scholar
Zelger, B, Cerio, R. Xanthogranuloma is the archetype of non‐Langerhans cell histiocytoses. Br J Dermatol. 2001;145(2):369–70.Google Scholar
Weitzman, S, Jaffe, R. Uncommon histiocytic disorders: the non-Langerhans cell histiocytoses. Pediatr Blood Cancer. 2005;45(3):256–64.Google Scholar
Ceppi, F, Abla, O. Juvenile xanthogranuloma and related non-LCH disorders. In: Abla, O, Janka, G, eds. Histiocytic disorders. Cham, Switzerland: Springer International; 2018: 293311.Google Scholar
Janssen, D, Harms, D. Juvenile xanthogranuloma in childhood and adolescence: a clinicopathologic study of 129 patients from the Kiel Pediatric Tumor Registry. Am J Surg Pathol. 2005;29(1):21–8.Google Scholar
Haynes, ESE, Guo, H, Jaffe, R, Picarsic, J. S100 immunohistochemistry in 65 localized juvenile xanthogranuloma family lesions: understanding staining patterns with clinicopathologic correlation. Presentation at the Spring 2017 Meeting of the Society for Pediatric Pathology, San Antonio, TX.Google Scholar
Haroche, J, Charlotte, F, Arnaud, L, von Deimling, A, Helias-Rodzewicz, Z, Hervier, B, et al. High prevalence of BRAF V600E mutations in Erdheim-Chester disease but not in other non-Langerhans cell histiocytoses. Blood. 2012;120(13):2700–3.Google Scholar
Picarsic, J, Pysher, T, Zhou, H, Fluchel, M, Pettit, T, Whitehead, M, et al. BRAF V600E mutation in juvenile xanthogranuloma family neoplasms of the central nervous system (CNS-JXG): a revised diagnostic algorithm to include pediatric Erdheim-Chester disease. Acta Neuropathol Commun. 2019;7(1):168.Google Scholar
Eissa, SS, Clay, MR, Santiago, T, Wu, G, Wang, L, Shulkin, BL, et al. Dasatinib induces a dramatic response in a child with refractory juvenile xanthogranuloma with a novel MRC1-PDGFRB fusion. Blood Adv. 2020;4(13):2991.Google Scholar
Lee, LH, Gasilina, A, Roychoudhury, J, Clark, J, McCormack, FX, Pressey, J, et al. Real-time genomic profiling of histiocytoses identifies early-kinase domain BRAF alterations while improving treatment outcomes. JCI Insight. 2017;2(3):e89473.Google Scholar
Hu, WK, Gilliam, AC, Wiersma, SR, Dahms, BB. Fatal congenital systemic juvenile xanthogranuloma with liver failure. Pediatr Dev Pathol. 2004;7(1):71–6.Google Scholar
Perez‐Becker, R, Szczepanowski, M, Leuschner, I, Janka, G, Gokel, M, Imschweiler, T, et al. An aggressive systemic juvenile xanthogranuloma clonally related to a preceding T‐cell acute lymphoblastic leukemia. Pediatr Blood Cancer. 2011;56(5):859–62.Google Scholar
Kemps, PG, Picarsic, J, Durham, BH, Hélias-Rodzewicz, Z, Hiemcke-Jiwa, L, van den Bos, C, et al. ALK-positive histiocytosis: a new clinicopathologic spectrum highlighting neurologic involvement and responses to ALK inhibition. Blood. 2022;139(2):256–80.Google Scholar
Jaffe, ES, Chan, JKC. Histiocytoses converge through common pathways. Blood. 2022;139(2):157–9.Google Scholar
Chan, JK, Lamant, L, Algar, E, Delsol, G, Tsang, WY, Lee, KC, et al. ALK+ histiocytosis: a novel type of systemic histiocytic proliferative disorder of early infancy. Blood. 2008;112(7):2965–8.Google Scholar
Huang, H, Gheorghe, G, North, PE, Suchi, M. Expanding the phenotype of ALK-positive histiocytosis: a report of 2 cases. Pediatr Dev Pathol. 2018;21(5):449–55.Google Scholar
Chang, KTE, Tay, AZE, Kuick, CH, Chen, H, Algar, E, Taubenheim, N, et al. ALK-positive histiocytosis: an expanded clinicopathologic spectrum and frequent presence of KIF5B-ALK fusion. Mod Pathol. 2019;32(5):598608.Google Scholar
Cuviello, A, Rice, J, Cohen, B, Zambidis, ET. Infant with a skin lesion and respiratory distress. BMJ Case Rep. 2018;2018:bcr–2018–224506.Google Scholar
Gupta, GK, Xi, L, Pack, SD, Jones, JB, Pittaluga, S, Raffeld, M, et al. ALK-positive histiocytosis with KIF5B-ALK fusion in an adult female. Haematologica. 2019;104(11):e534.Google Scholar
Lucas, C-HG, Gilani, A, Solomon, DA, Liang, X, Maher, OM, Chamyan, G, et al. ALK-positive histiocytosis with KIF5B-ALK fusion in the central nervous system. Acta neuropathol. 2019;138(2):335–7.Google Scholar
Wolter, NE, Ngan, B, Whitlock, JA, Dickson, BC, Propst, EJ. Atypical juvenile histiocytosis with novel KIF5B-ALK gene fusion mimicking subglottic hemangioma. Int J Pediatr Otorhinolaryngol. 2019;126:109585.Google Scholar
Sarah, M, Zofia, H-R, Fleur, C-A, Frédéric, C, Sylvie, F, Nathalie, T, et al. Highly sensitive methods are required to detect mutations in histiocytoses. Haematologica. 2019;104(3):e97–e9.Google Scholar
Destombes, P. [Adenitis with lipid excess, in children or young adults, seen in the Antilles and in Mali. (4 cases)]. Bull Soc Pathol Exot Filiales. 1965;58(6):1169–75.Google Scholar
Rosai, J, Dorfman, RF. Sinus histiocytosis with massive lymphadenopathy.A newly recognized benign clinicopathological entity. Arch Pathol. 1969;87(1):6370.Google Scholar
Abla, O, Picarsic, J. Pathology of histiocytic disorders and neoplasms and related disorders. In: Abla, O, Janka, G, eds. Histiocytic disorders. Springer; 2017:339–60.Google Scholar
Morgan, NV, Morris, MR, Cangul, H, Gleeson, D, Straatman-Iwanowska, A, Davies, N, et al. Mutations in SLC29A3, encoding an equilibrative nucleoside transporter ENT3, cause a familial histiocytosis syndrome (Faisalabad histiocytosis) and familial Rosai-Dorfman disease. PLoS Genet. 2010;6(2):e1000833.Google Scholar
Maric, I, Pittaluga, S, Dale, JK, Niemela, JE, Delsol, G, Diment, J, et al. Histologic features of sinus histiocytosis with massive lymphadenopathy in patients with autoimmune lymphoproliferative syndrome. Am J Surg Pathol. 2005;29(7):903–11.Google Scholar
Mastropolo, R, Close, A, Allen, SW, McClain, KL, Maurer, S, Picarsic, J. BRAF-V600E-mutated Rosai-Dorfman-Destombes disease and Langerhans cell histiocytosis with response to BRAF inhibitor. Blood Adv. 2019;3(12):1848–53.Google Scholar
Fatobene, G, Haroche, J, Helias-Rodzwicz, Z, Charlotte, F, Taly, V, Ferreira, AM, et al. BRAF V600E mutation detected in a case of Rosai-Dorfman disease. Haematologica. 2018;103(8):e377–e9.Google Scholar
Richardson, TE, Wachsmann, M, Oliver, D, Abedin, Z, Ye, D, Burns, DK, et al. BRAF mutation leading to central nervous system Rosai-Dorfman disease. Ann Neurol. 2018;84(1):147–52.Google Scholar
Moore, JC, Zhao, X, Nelson, EL. Concomitant sinus histiocytosis with massive lymphadenopathy (Rosai-Dorfman disease) and diffuse large B-cell lymphoma: a case report. J Med Case Rep. 2008;2:70.Google Scholar
Johnson, WT, Patel, P, Hernandez, A, Grandinetti, LM, Huen, AC, Marks, S, et al. Langerhans cell histiocytosis and Erdheim-Chester disease, both with cutaneous presentations, and papillary thyroid carcinoma all harboring the BRAF(V600E) mutation. J Cutan Pathol. 2016;43(3):270–5.Google Scholar
Hervier, B, Haroche, J, Arnaud, L, Charlotte, F, Donadieu, J, Neel, A, et al. Association of both Langerhans cell histiocytosis and Erdheim-Chester disease linked to the BRAFV600E mutation: a multicenter study of 23 cases. Blood. 2014;124(7):1119–26.Google Scholar
Milne, P, Bigley, V, Bacon, CM, Neel, A, McGovern, N, Bomken, S, et al. Hematopoietic origin of Langerhans cell histiocytosis and Erdheim Chester disease in adults. Blood. 2017;130(2):167–75.Google Scholar
Berres, ML, Merad, M, Allen, CE. Progress in understanding the pathogenesis of Langerhans cell histiocytosis: back to histiocytosis X? Br J Haematol. 2015;169(1):313.Google Scholar
Egan, C, Lack, J, Skarshaug, S, Pham, TA, Abdullaev, Z, Xi, L, et al. The mutational landscape of histiocytic sarcoma associated with lymphoid malignancy. Mod Pathol. 2020:34(2):336347.Google Scholar
Castro, EC, Blazquez, C, Boyd, J, Correa, H, de Chadarevian, JP, Felgar, RE, et al. Clinicopathologic features of histiocytic lesions following ALL, with a review of the literature. Pediatr Dev Pathol. 2010;13(3):225–37.Google Scholar
Howard, JE, Dwivedi, RC, Masterson, L, Jani, P. Langerhans cell sarcoma: a systematic review. Cancer Treat Rev. 2015;41(4):320–31.Google Scholar
Zwerdling, T, Won, E, Shane, L, Javahara, R, Jaffe, R. Langerhans cell sarcoma: case report and review of world literature. J Pediatr Hematol Oncol. 2014;36(6):419–25.Google Scholar
Pileri, SA, Grogan, TM, Harris, NL, Banks, P, Campo, E, Chan, JK, et al. Tumours of histiocytes and accessory dendritic cells: an immunohistochemical approach to classification from the International Lymphoma Study Group based on 61 cases. Histopathology. 2002;41(1):129.Google Scholar
Swerdlow, SH, Campo, E, Harris, NL, Jaffe, ES, Pileri, SA, Stein, H, et al., eds. WHO classification of tumours of haematopoietic and lymphoid tissues. Rev. 4th ed. Lyon, France: IARC; 2017.Google Scholar
Singh, R, Keen, CE, Stone, C, Sarsfield, P. Langerhans cell sarcoma: a case report demonstrating morphological and immunophenotypical variability within a single lesion. Case Rep Pathol. 2017;2017:9842605.Google Scholar
Karai, LJ, Sanik, E, Ricotti, CA, Susa, J, Sinkre, P, Aleodor, AA. Langerhans cell sarcoma with lineage infidelity/plasticity: a diagnostic challenge and insight into the pathobiology of the disease. Am J Dermatopathol. 2015;37(11):854–61.Google Scholar
Xu, Z, Padmore, R, Faught, C, Duffet, L, Burns, BF. Langerhans cell sarcoma with an aberrant cytoplasmic CD3 expression. Diagn Pathol. 2012;7:128.Google Scholar
Jacobsen, E, Abla, O, Visser, J. Malignant histiocytoses. In: Histiocytic disorders. Springer; 2018: 361–81.Google Scholar
Swerdlow, SH, Campo, E, Pileri, SA, Harris, NL, Stein, H, Siebert, R, et al. The 2016 revision of the World Health Organization (WHO) classification of lymphoid neoplasms. Blood. 2016;127(20):2391–405.Google Scholar
Shanmugam, V, Sholl, L, Fletcher, CD, Hornick, JL, eds. RAS/MAPK pathway activation defines a common molecular subtype of histiocytic sarcoma. Laboratory investigation. New York: Nature Publishing Group; 2018.Google Scholar
Heath, JL, Burgett, SE, Gaca, AM, Jaffe, R, Wechsler, DS. Successful treatment of pediatric histiocytic sarcoma using abbreviated high-risk leukemia chemotherapy. Pediatr Blood Cancer. 2014;61(10):1874–6.Google Scholar
Porter, DW, Gupte, GL, Brown, RM, Spray, C, English, MW, DeGoyet, Jd, et al. Histiocytic sarcoma with interdigitating dendritic cell differentiation. J Pediatr. Hematol. Oncol. 2004;26(12):827–30.Google Scholar
Kordes, M, Roring, M, Heining, C, Braun, S, Hutter, B, Richter, D, et al. Cooperation of BRAF(F595L) and mutant HRAS in histiocytic sarcoma provides new insights into oncogenic BRAF signaling. Leukemia. 2016;30(4):937–46.Google Scholar
Chakraborty, R, Abdel-Wahab, O, Durham, BH. MAP-kinase-driven hematopoietic neoplasms: a decade of progress in the molecular age. Cold Spring Harb Perspect Med. 2020;11(5):a034892.Google Scholar
Idbaih, A, Mokhtari, K, Emile, JF, Galanaud, D, Belaid, H, de Bernard, S, et al. Dramatic response of a BRAF V600E-mutated primary CNS histiocytic sarcoma to vemurafenib. Neurology. 2014;83(16):1478–80.Google Scholar
Xerri, L, Adélaïde, J, Popovici, C, Garnier, S, Guille, A, Mescam-Mancini, L, et al. CDKN2A/B deletion and double-hit mutations of the MAPK pathway underlie the aggressive behavior of Langerhans cell tumors. Am J Surg Pathol. 2018;42(2):150–9.Google Scholar
Liu, Q, Tomaszewicz, K, Hutchinson, L, Hornick, JL, Woda, B, Yu, H. Somatic mutations in histiocytic sarcoma identified by next generation sequencing. Virchows Arch. 2016;469(2):233-41.Google Scholar
Go, H, Jeon, YK, Huh, J, Choi, SJ, Choi, YD, Cha, HJ, et al. Frequent detection of BRAF(V600E) mutations in histiocytic and dendritic cell neoplasms. Histopathology. 2014;65(2):261–72.Google Scholar
Ansari, J, Naqash, AR, Munker, R, El-Osta, H, Master, S, Cotelingam, JD, et al. Histiocytic sarcoma as a secondary malignancy: pathobiology, diagnosis, and treatment. Eur J Haematol. 2016;97(1):916.Google Scholar
West, DS, Dogan, A, Quint, PS, Tricker-Klar, ML, Porcher, JC, Ketterling, RP, et al. Clonally related follicular lymphomas and Langerhans cell neoplasms: expanding the spectrum of transdifferentiation. Am J Surg Pathol. 2013;37(7):978–86.Google Scholar
Choi, SM, Andea, AA, Wang, M, Behdad, A, Shao, L, Zhang, Y, et al. KRAS mutation in secondary malignant histiocytosis arising from low grade follicular lymphoma. Diagn Pathol. 2018;13(1):78.Google Scholar
Feldman, AL. Clonal relationships between malignant lymphomas and histiocytic/dendritic cell tumors. Surg Pathol Clin. 2013;6(4):619–29.Google Scholar
Kumar, R, Khan, SP, Joshi, DD, Shaw, GR, Ketterling, RP, Feldman, AL. Pediatric histiocytic sarcoma clonally related to precursor B-cell acute lymphoblastic leukemia with homozygous deletion of CDKN2A encoding p16(INK4A). Pediatr Blood Cancer. 2010;56(2):307–10.Google Scholar
Rodig, SJ, Payne, EG, Degar, BA, Rollins, B, Feldman, AL, Jaffe, ES, et al. Aggressive Langerhans cell histiocytosis following T-ALL: clonally related neoplasms with persistent expression of constitutively active NOTCH1. Am J Hematol. 2008;83(2):116–21.Google Scholar
Feldman, AL, Berthold, F, Arceci, RJ, Abramowsky, C, Shehata, BM, Mann, KP, et al. Clonal relationship between precursor T-lymphoblastic leukaemia/lymphoma and Langerhans-cell histiocytosis. Lancet Oncol. 2005;6(6):435–7.Google Scholar
Shao, H, Xi, L, Raffeld, M, Feldman, AL, Ketterling, RP, Knudson, R, et al. Clonally related histiocytic/dendritic cell sarcoma and chronic lymphocytic leukemia/small lymphocytic lymphoma: a study of seven cases. Mod Pathol. 2011;24(11):1421–32.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×