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

Histiocytic sarcoma

Histiocytic sarcoma
Author:
Eric Jacobsen, MD
Section Editor:
Arnold S Freedman, MD
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Jan 2024.
This topic last updated: Jan 31, 2022.

INTRODUCTION — Histiocytic sarcoma (HS) is an extremely rare non-Langerhans histiocyte disorder of unknown cause that most commonly presents with symptoms due to unifocal or multifocal extranodal tumors. HS may occur as a sporadic illness or may be clonally related to a separate synchronous or metachronous hematologic malignancy, such as follicular lymphoma or acute lymphoblastic leukemia.

Histiocytic disorders are thought to be derived from mononuclear phagocytic cells (macrophages and dendritic cells) or histiocytes. This group has generally been divided into Langerhans cell histiocytosis and non-Langerhans histiocytosis. Langerhans cell histiocytosis is so named for its presumed derivation from the Langerhans cells, which are specialized dendritic cells found in the skin and mucosa. In contrast, non-Langerhans histiocytoses are thought to be derived from the monocyte-macrophage lineage.

The epidemiology, clinical manifestations, pathologic features, diagnosis, and management of HS will be presented here. The diagnosis and management of Langerhans cell histiocytosis and other non-Langerhans histiocytic disorders are presented separately. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis" and "Pulmonary Langerhans cell histiocytosis" and "Erdheim-Chester disease".)

EPIDEMIOLOGY — HS is an extremely rare disease with only a few hundred cases reported in the literature. HS has been diagnosed in all age groups, but is most common in adults. A study of 159 cases of HS (99 men, 60 women) from the US Surveillance, Epidemiology, and End Results (SEER) database reported a median age at diagnosis of 63 years (range 18 to 96 years) [1]. The incidence was lower in African Americans than in White Americans (incidence ratio 0.27) and higher in men (female:male incidence ratio 0.49). In two other case series, the median ages at diagnosis were 46 and 55 years [2,3].

There are no known environmental or hereditary genetic factors predisposing to the development of HS. HS can occur as an isolated disease or in the context of other hematologic neoplasms, such as follicular lymphoma, myelodysplasia, or acute lymphoblastic leukemia [1,3]. When HS occurs in the context of another hematologic malignancy, the two entities are often clonally related [4,5]. Of importance, tumors with pathologic features consistent with HS occurring in the context of acute monocytic leukemia are not considered HS.

PATHOGENESIS — The pathogenesis of HS remains unclear. HS is not a true sarcoma but is derived from cells of the monocytes/macrophage system. HS has been described in association with a number of hematologic neoplasms, most commonly acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and follicular lymphoma (FL). In addition, cases have been reported in patients with mediastinal germ cell tumors, such as malignant teratomas, suggesting a possible derivation of such tumors from pluripotent germ cells [6].

Attempts to establish clonality have had varied results, and although occasional cytogenetic aberrations have been identified, none are characteristic:

In some cases of HS associated with lymphoid neoplasms, the HS has been shown to originate from the same clone that gave rise to the lymphoid neoplasm [4,5].

In a landmark study of eight patients with both FL and HS (seven patients) or interdigitating dendritic cell sarcoma (one patient), all eight possessed the t(14;18) either by fluorescence in situ hybridization (FISH) or by polymerase chain reaction (PCR) [7]. Five HS lesions were metachronous, developing two months to 12 years after the follicular lymphoma, while the FL and HS were synchronous in three cases.

In a large study of sporadic HS, 9 of 23 cases (39 percent) demonstrated clonal immunoglobulin (Ig) heavy chain (with or without Ig kappa) gene rearrangements, while two (9 percent) cases showed only clonal Ig kappa gene rearrangements [8]. One case showed t(14;18) by PCR and FISH analysis showed the resulting IGH/BCL2 fusion in the neoplastic histiocytes. All cases of Ig heavy chain and Ig kappa positive HS were negative for B cell-associated transcription factors such as PAX5 and BOB.1, though four of seven were positive for Oct2.

Human and mouse HS tumors have demonstrated genetic or epigenetic inactivation of PTEN, INK4A (p16), and ARF (p14), but how or why these changes occur remains unclear [9].

BRAF V600E mutations are well described in other histiocyte/dendritic cell neoplasms such as Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease, and there are reports of HS harboring the BRAF V600E mutation [10-12]. Other mutations in the MAP2K1 pathway that may be targeted by MEK inhibitors have also been described [13,14]. Given the potential therapeutic implications and conflicting results from published studies, we test tumors from all patients with HS for BRAF mutation by PCR and/or immunohistochemistry.

CLINICAL MANIFESTATIONS — The clinical presentation of HS varies depending upon the organs involved. Most patients present with symptoms due to unifocal or multifocal extranodal disease, most commonly involving the intestinal tract, skin, and soft tissues [15]. However, there is no characteristic pattern of organ system involvement and any organ may be involved including bone, lymph nodes, skin, liver, spleen, lung and the central nervous system. Solitary involvement of the lymph nodes is seen in less than 20 percent of cases [3].

Organ involvement may be asymptomatic and discovered at the time of radiographic imaging. More commonly, patients present with a palpable mass lesion, symptoms related to compression of surrounding organs (eg, intestinal obstruction), or systemic complaints (eg, fevers and weight loss). Skin involvement has numerous potential manifestations ranging from rash to tumorous growths [15]. Laboratory abnormalities depend on the organ system affected. Cytopenias are seen in approximately one-third of cases, a minority of which will demonstrate hemophagocytosis on bone marrow biopsy [3,16].

PATHOLOGIC FEATURES — Biopsies of involved tissue demonstrate an infiltrative process with a diffuse growth pattern and effacement of the normal architecture [15]. When involved, the lymph nodes, bone marrow, and spleen demonstrate sinusoidal infiltration and parenchymal involvement. Skin biopsies do not demonstrate epidermotropism.

The tumor is primarily comprised of non-cohesive large cells (eg, nuclei at least twice the size of a small lymphocyte and larger than the nucleus of a tissue macrophage) that are polygonal or ovoid with focal spindling (picture 1) [3,17]. The cells have abundant eosinophilic cytoplasm that may contain fine vacuoles. The nuclei are pleomorphic and slightly eccentric with moderately dispersed chromatin and one or more small, but prominent nucleoli. Occasionally, the neoplastic cells have a foamy (xanthomatous) appearance.

Hemophagocytosis, tumor giant cells, or a prominent inflammatory infiltrate may be present [18]. Tissue necrosis is not prominent, but may be seen focally in a minority of cases. Occasionally a prominent inflammatory infiltrate obscures the less abundant tumor cells. Birbeck granules (a seminal finding in Langerhans cell histiocytosis) and cellular junctions are absent on electron microscopy.

On immunohistochemistry, the cells typically express CD68, lysozyme, CD4 and CD163, but do not express specific T and B cell markers, myeloid cell markers (eg, myeloperoxidase, CD33), CD1a, S100 protein or epithelial markers (picture 2) [3,19,20]. Staining for lysozyme is usually granular with accentuation of the Golgi [3].

T cell receptor (TCR) and immunoglobulin genes are usually germline, but clonal rearrangements of antigen receptor genes have been identified in some cases. (See 'Pathogenesis' above.)

DIAGNOSIS AND DIFFERENTIAL DIAGNOSIS — HS is diagnosed based upon the pathologic evaluation of involved tissue interpreted within the clinical context [17]. HS can be difficult to diagnose since it is a very rare disease that can affect many organ systems. An excisional or incisional biopsy of affected tissue is generally preferred, when possible, over a fine needle aspirate or core needle biopsy.

HS must be distinguished from other histiocytic and dendritic cell disorders, metastatic solid or hematopoietic neoplasms, primary familial lymphohistiocytic disorders, and acquired causes of hemophagocytic macrophage activation syndromes. Of importance, tumors with pathologic features consistent with HS occurring in the context of acute monocytic leukemia are not considered HS.

Langerhans cell histiocytosis – Langerhans cell histiocytosis (LCH) and HS are both histiocytic diseases that can involve multiple sites. The two entities can usually be distinguished by morphologic and immunohistochemical evaluation. Unlike Langerhans cell histiocytosis, HS tumor cells do not express CD1a or S100. Birbeck granules, a seminal finding in Langerhans cell histiocytosis, are absent in HS. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

Hemophagocytic lymphohistiocytosis – Hemophagocytic lymphohistiocytosis and the related macrophage activation syndrome are systemic disorders that demonstrate tissue infiltration by non-neoplastic histiocytes. While both disorders may demonstrate hemophagocytosis, phagocytic activity is much more common in hemophagocytic lymphohistiocytosis. (See "Treatment and prognosis of hemophagocytic lymphohistiocytosis".)

Lymphoma – Morphologically, HS resembles diffuse large cell lymphoma or anaplastic large cell lymphoma. However, these entities can usually be easily distinguished from HS by immunohistochemistry. HS does not express B or T cell markers (eg, myeloperoxidase, CD33) and T cell receptor and immunoglobulin genes are usually germline. (See "Epidemiology, clinical manifestations, pathologic features, and diagnosis of diffuse large B cell lymphoma" and "Clinical manifestations, pathologic features, and diagnosis of systemic anaplastic large cell lymphoma (sALCL)".)

Metastatic carcinoma – Morphologically, lymph node involvement with carcinoma may resemble HS in that both tumors are comprised of large atypical cells. Carcinoma can be readily distinguished from HS by the demonstration of cytokeratin upon staining.

Metastatic melanoma – Both melanoma and HS have large atypical cells and can involve multiple organ systems. However, unlike HS, melanoma is positive for S100, HMB-45, and/or Melan A. (See "Pathologic characteristics of melanoma".)

STAGING — To best treat patients with HS, the initial evaluation must confirm the histologic diagnosis, the extent and sites of disease, and the performance status of the patient. In addition to a history and physical examination, it is our practice to perform the following pretreatment studies in patients with HS:

Laboratory studies include a complete blood count, serum electrolytes, and chemistries with liver and renal function.

Computed tomography (CT) or a combined positron emission tomography (PET)/CT to document organ involvement. HS appears to be FDG-avid, at least in small case series [21].

Bone marrow biopsy for patients with cytopenias or findings suggestive of hemophagocytosis.

MANAGEMENT — There is no standardized therapy for HS and patients should be encouraged to enroll in a clinical trial if one is available. Outside of a clinical trial, treatment options include surgery, radiotherapy, and systemic chemotherapy. The choice among these options is primarily made based upon the extent of disease. Multisystem disease and or a solitary tumor greater than 3.5 cm seem to carry the worst prognosis [2].

Unifocal disease — The ideal treatment regimen for patients with unifocal HS is unknown. Data from the published literature are limited to small case series and single-case reports. In addition, the natural history of HS is largely unknown and is likely variable. For patients with unifocal HS, we suggest surgical resection plus adjuvant radiation therapy. Though the optimal radiation dose and field are unknown, we favor involved field radiation therapy at 45 to 50 Gy. There is no clear role for adjuvant chemotherapy in this setting.

The largest case series included 14 patients presenting with unifocal extranodal HS treated at a single institution [2]. Surgical resection data was available for 12 patients: six tumors were excised with wide margins, four were excised with minimal margins (one with positive margins), and two were biopsied, but not excised. The following describes the variability of treatment and outcomes:

Five patients were treated with surgical resection alone. Of these, two developed distant spread within three to four months. Two patients were alive with no sign of recurrence at short follow-up. The fifth patient developed local recurrence at six months that was treated with further resection and radiation therapy and had not recurred again at 11 years of follow-up.

Three patients were treated with surgical resection plus adjuvant radiation therapy. None have recurred locally. One developed distant recurrences at 17 and 26 months that were treated with further resection. All three patients were alive with no evidence of disease at a median follow-up of 22 months.

Six patients were treated with adjuvant chemotherapy (primarily CHOP: cyclophosphamide, doxorubicin, vincristine, and prednisone). Two patients developed distant spread within weeks, one of whom was successfully treated with further chemotherapy. Two were alive with no evidence of disease at median of 16 months of follow-up. Two patients did not have adequate follow-up.

Based upon this data and our clinical experience, we prefer surgical resection with adjuvant radiation therapy. This approach appears to provide good local control while minimizing toxicity. Patients who undergo an initial surgical resection and then experience an isolated site of recurrence may be considered for serial resection and/or radiation therapy.

Multisystem disease — Patients with multisystem involvement appear to have a more aggressive clinical course than those with unifocal disease. Given the rarity of this disease, there are no large prospective trials. Data regarding treatment are limited to small case series and single-case reports. For patients with multisystem involvement of HS, we suggest the use of combination chemotherapy. Surgical intervention is limited to diagnostic biopsies and the management of complications such as perforation, obstruction, or intractable bleeding. There is no clear role for adjuvant radiation therapy in this setting.

The ideal chemotherapy regimen for HS is unknown. Most clinicians use regimens designed for patients with clinically aggressive lymphomas, such as diffuse large B cell lymphoma. The two most commonly used regimens are:

Ifosfamide, carboplatin, and etoposide with mesna (ICE) – Toxicity data come from the use of ICE in other malignancies [22]. Hematologic toxicity is universal with 35 percent of patients requiring transfusion of blood products. Severe (grade 3/4) nonhematologic side effects include infection (23 percent) and nephrotoxicity (1 percent).

Cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) – Toxicity data come from the use of CHOP in other malignancies [23]. Hematologic toxicity is common, but less than 20 percent require transfusion of blood products. The most common severe (grade 3/4) nonhematologic toxicities are infection (20 percent), lung toxicity (10 percent), and neurologic toxicity (9 percent).  

These two regimens have not been compared directly for the treatment of HS. For patients with a good performance status, we prefer six cycles of ICE chemotherapy. Six to eight cycles of CHOP chemotherapy is an acceptable alternative and is generally preferred for older patients who are unlikely to tolerate ICE [2]. This preference for ICE over CHOP is primarily because ICE is a more aggressive chemotherapy regimen than CHOP and would theoretically have a greater potential for response in HS (an entity that is often resistant to chemotherapy). Patients who cannot tolerate multiagent chemotherapy (ie, ICE or CHOP) can be treated with single agent vinblastine, etoposide, and/or prednisone modeled after the Histiocyte Society treatment protocol for Langerhans histiocytosis. Appropriate candidates with advanced stage disease who achieve remission with front-line treatment should be considered for allogeneic stem cell transplantation, given the high recurrence rate after chemotherapy alone. (See "Clinical manifestations, pathologic features, and diagnosis of Langerhans cell histiocytosis".)

BRAF inhibitors have demonstrated efficacy in Langerhans cell histiocytosis (LCH) and Erdheim-Chester disease (ECD) harboring the BRAF V600E mutation; responses BRAF inhibitors have been reported in HS, but their activity in HS is not well documented [24-26]. Inhibitors of BRAF or MEK (for MAP2K1 pathway mutations) should not be given as first-line therapy in HS outside of a clinical trial. Use of these inhibitors in relapsed disease is discussed below. (See 'Relapsed disease' below.)

PATIENT FOLLOW-UP — Patients should be clinically assessed a minimum of every three to six months or more frequently as symptoms and organ dysfunction require. The frequency of imaging is dictated by baseline disease status, organ involvement, and requirement for ongoing treatment. Affected organs should be imaged every three to six months until stability is documented.

RELAPSED DISEASE — Patients who undergo an initial surgical resection and then experience an isolated site of recurrence may be considered for serial resection and/or radiation therapy. Patients with relapsed or refractory multisystem disease are generally treated with systemic chemotherapy with plans to proceed to allogeneic hematopoietic cell transplantation (HCT) if they achieve a remission and allogeneic transplant was not previously performed. Although there is a paucity of data to support the use of allogeneic HCT in this setting, conventional chemotherapy regimens provide only transient disease control for most patients with relapsed or refractory HS. Allogeneic HCT provides additional intensive chemotherapy and a potential graft-versus-tumor effect. Patients who are not candidates for allogeneic HCT may respond to high dose chemotherapy followed by autologous HCT rescue [27,28]. Radiation therapy may be used for local palliation.

The ideal chemotherapy regimen for relapsed or refractory HS is unknown, but regimens that contain non-cross resistant drugs are generally used. As such, patients initially treated with ICE may be considered for treatment with CHOP. Patients with relapsed/refractory HS that harbors the BRAF V600E mutation after systemic therapy should be treated with vemurafenib or dabrafenib, preferably in the context of a clinical trial; these agents have demonstrated efficacy against Langerhans cell histiocytosis and Erdheim-Chester disease. We generally begin with vemurafenib 480 mg twice daily (rather than 960 mg twice daily used in other settings) and the dose can be adjusted based upon response and toxicity. Patients with mutations in the MAP2K1 pathway may be sensitive to MEK inhibitors. There are case reports of thalidomide [28,29] and alemtuzumab [30] showing activity. HS frequently overexpresses PD-L1 and there are case reports of responses to PD-1 inhibitors [31-33]. Ideally such treatment should be used in the context of a clinical trial.

CLINICAL TRIALS — Often there is no better therapy to offer a patient than enrollment in a well-designed, scientifically valid, peer-reviewed clinical trial. Additional information and instructions for referring a patient to an appropriate research center can be obtained from the National Cancer Institute or from the Histiocyte Society.

SUMMARY AND RECOMMENDATIONS

Histiocytic sarcoma (HS) is a rare histiocyte disorder that can present as unifocal or multifocal disease affecting nearly any organ system. (See 'Epidemiology' above and 'Pathogenesis' above.)

Clinical presentation varies depending upon the sites of involvement. Most patients present with symptoms due to unifocal or multifocal extranodal disease, most commonly involving the intestinal tract, skin, and soft tissues. (See 'Clinical manifestations' above.)

Diagnosis – HS is diagnosed based upon the pathologic evaluation of involved tissue interpreted within the clinical context. HS can be difficult to diagnose since it is a very rare disease and can affect many organ systems. An excisional or incisional biopsy of an affected organ is generally preferred, when possible. (See 'Pathologic features' above and 'Diagnosis and differential diagnosis' above.)

Management – There is no standard treatment regimen for patients with HS and patients should be encouraged to enroll in a clinical trial. We use the following approach for the management of patients outside of a clinical trial (see 'Clinical trials' above):

Unifocal disease – For patients with unifocal disease we suggest surgical excision with adjuvant radiotherapy, if possible, depending on location and extent of disease (Grade 2C). If radiation is not feasible or would be too toxic, post-operative observation is reasonable. Patients may have serial unifocal recurrences over time and repeat surgical excision and/or radiotherapy is reasonable in these instances. (See 'Unifocal disease' above.)

Multifocal disease – For patients with multifocal disease, we suggest aggressive multiagent chemotherapy with six cycles of ifosfamide, mesna, carboplatin, and etoposide (ICE) (Grade 2C). Cyclophosphamide, doxorubicin, vincristine, prednisone (CHOP) is an acceptable alternative for those who cannot tolerate ICE. Glucocorticoids, single agent vinblastine, and single agent etoposide are generally reserved for patients who cannot tolerate combination chemotherapy. (See 'Multisystem disease' above.)

Allogeneic hematopoietic cell transplantation (HCT) should be considered for medically eligible patients with advanced stage disease who achieve remission.

Relapsed/refractory HS – Patients with relapsed or refractory disease should be considered for treatment in a clinical trial, when available. Outside of a clinical trial, systemic therapy with non-cross resistant drugs, followed by allogeneic HCT should be offered to medically eligible patients. Alternatives include inhibitors of BRAF or MEK (for patients with targetable mutations), thalidomide, alemtuzumab, and PD-1 inhibitors. (See 'Relapsed disease' above.)

  1. Kommalapati A, Tella SH, Durkin M, et al. Histiocytic sarcoma: a population-based analysis of incidence, demographic disparities, and long-term outcomes. Blood 2018; 131:265.
  2. Hornick JL, Jaffe ES, Fletcher CD. Extranodal histiocytic sarcoma: clinicopathologic analysis of 14 cases of a rare epithelioid malignancy. Am J Surg Pathol 2004; 28:1133.
  3. Pileri SA, Grogan TM, Harris NL, 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.
  4. Castro EC, Blazquez C, Boyd J, et al. Clinicopathologic features of histiocytic lesions following ALL, with a review of the literature. Pediatr Dev Pathol 2010; 13:225.
  5. Wang E, Hutchinson CB, Huang Q, et al. Histiocytic sarcoma arising in indolent small B-cell lymphoma: report of two cases with molecular/genetic evidence suggestive of a 'transdifferentiation' during the clonal evolution. Leuk Lymphoma 2010; 51:802.
  6. deMent SH. Association between mediastinal germ cell tumors and hematologic malignancies: an update. Hum Pathol 1990; 21:699.
  7. Feldman AL, Arber DA, Pittaluga S, et al. Clonally related follicular lymphomas and histiocytic/dendritic cell sarcomas: evidence for transdifferentiation of the follicular lymphoma clone. Blood 2008; 111:5433.
  8. Chen W, Lau SK, Fong D, et al. High frequency of clonal immunoglobulin receptor gene rearrangements in sporadic histiocytic/dendritic cell sarcomas. Am J Surg Pathol 2009; 33:863.
  9. Carrasco DR, Fenton T, Sukhdeo K, et al. The PTEN and INK4A/ARF tumor suppressors maintain myelolymphoid homeostasis and cooperate to constrain histiocytic sarcoma development in humans. Cancer Cell 2006; 9:379.
  10. Go H, Jeon YK, Huh J, et al. Frequent detection of BRAF(V600E) mutations in histiocytic and dendritic cell neoplasms. Histopathology 2014; 65:261.
  11. Haroche J, Charlotte F, Arnaud L, et al. High prevalence of BRAF V600E mutations in Erdheim-Chester disease but not in other non-Langerhans cell histiocytoses. Blood 2012; 120:2700.
  12. Liu Q, Tomaszewicz K, Hutchinson L, et al. Somatic mutations in histiocytic sarcoma identified by next generation sequencing. Virchows Arch 2016; 469:233.
  13. Gounder MM, Solit DB, Tap WD. Trametinib in Histiocytic Sarcoma with an Activating MAP2K1 (MEK1) Mutation. N Engl J Med 2018; 378:1945.
  14. Shanmugam V, Griffin GK, Jacobsen ED, et al. Identification of diverse activating mutations of the RAS-MAPK pathway in histiocytic sarcoma. Mod Pathol 2019; 32:830.
  15. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues, Swerdlow SH, Campo E, Harris NL, et al. (Eds), IARC Press, Lyon 2008.
  16. Oka K, Nakamine H, Maeda K, et al. Primary histiocytic sarcoma of the spleen associated with hemophagocytosis. Int J Hematol 2008; 87:405.
  17. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016; 127:2375.
  18. Vos JA, Abbondanzo SL, Barekman CL, et al. Histiocytic sarcoma: a study of five cases including the histiocyte marker CD163. Mod Pathol 2005; 18:693.
  19. Yoshida C, Takeuchi M. Histiocytic sarcoma: identification of its histiocytic origin using immunohistochemistry. Intern Med 2008; 47:165.
  20. Mikami M, Sadahira Y, Suetsugu Y, et al. Monocyte/Macrophage-specific marker CD163+ histiocytic sarcoma: case report with clinical, morphologic, immunohistochemical, and molecular genetic studies. Int J Hematol 2004; 80:365.
  21. Yaman E, Ozturk B, Erdem O, et al. Histiocytic sarcoma: PET-CT evaluation of a rare entity. Ann Nucl Med 2008; 22:715.
  22. Gisselbrecht C, Glass B, Mounier N, et al. Salvage regimens with autologous transplantation for relapsed large B-cell lymphoma in the rituximab era. J Clin Oncol 2010; 28:4184.
  23. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma. N Engl J Med 2002; 346:235.
  24. Haroche J, Cohen-Aubart F, Emile JF, et al. Reproducible and sustained efficacy of targeted therapy with vemurafenib in patients with BRAF(V600E)-mutated Erdheim-Chester disease. J Clin Oncol 2015; 33:411.
  25. Haroche J, Cohen-Aubart F, Emile JF, et al. Dramatic efficacy of vemurafenib in both multisystemic and refractory Erdheim-Chester disease and Langerhans cell histiocytosis harboring the BRAF V600E mutation. Blood 2013; 121:1495.
  26. Idbaih A, Mokhtari K, Emile JF, et al. Dramatic response of a BRAF V600E-mutated primary CNS histiocytic sarcoma to vemurafenib. Neurology 2014; 83:1478.
  27. Mainardi C, D'Amore ES, Pillon M, et al. A case of resistant pediatric histiocytic sarcoma successfully treated with chemo-radiotherapy and autologous peripheral blood stem cell transplant. Leuk Lymphoma 2011; 52:1367.
  28. Gergis U, Dax H, Ritchie E, et al. Autologous hematopoietic stem-cell transplantation in combination with thalidomide as treatment for histiocytic sarcoma: a case report and review of the literature. J Clin Oncol 2011; 29:e251.
  29. Abidi MH, Tove I, Ibrahim RB, et al. Thalidomide for the treatment of histiocytic sarcoma after hematopoietic stem cell transplant. Am J Hematol 2007; 82:932.
  30. Shukla N, Kobos R, Renaud T, et al. Successful treatment of refractory metastatic histiocytic sarcoma with alemtuzumab. Cancer 2012; 118:3719.
  31. Gatalica Z, Bilalovic N, Palazzo JP, et al. Disseminated histiocytoses biomarkers beyond BRAFV600E: frequent expression of PD-L1. Oncotarget 2015; 6:19819.
  32. Xu J, Sun HH, Fletcher CD, et al. Expression of Programmed Cell Death 1 Ligands (PD-L1 and PD-L2) in Histiocytic and Dendritic Cell Disorders. Am J Surg Pathol 2016; 40:443.
  33. Bose S, Robles J, McCall CM, et al. Favorable response to nivolumab in a young adult patient with metastatic histiocytic sarcoma. Pediatr Blood Cancer 2019; 66:e27491.
Topic 16411 Version 27.0

References

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