Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
1.
Hum Pathol ; 147: 101-113, 2024 May.
Article in English | MEDLINE | ID: mdl-38280658

ABSTRACT

The WHO Classification of Soft Tissue and Bone Tumours currently recognizes four categories of undifferentiated small round cell sarcoma: Ewing sarcoma, round cell sarcoma with EWSR1-non-ETS fusions including NFATc2 and PATZ1, CIC-rearranged sarcoma, and sarcoma with BCOR genetic alterations. These neoplasms frequently pose significant diagnostic challenges due to rarity and overlapping morphologic and immunohistochemical findings. Further, molecular testing, with accompanying pitfalls, may be needed to establish a definitive diagnosis. This review summarizes the clinical, histologic, immunohistochemical, and molecular features of these neoplasms. In addition, differential diagnosis and areas of uncertainty and ongoing investigation are discussed.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Sarcoma, Ewing , Sarcoma, Small Cell , World Health Organization , Humans , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Sarcoma, Ewing/classification , Sarcoma, Ewing/chemistry , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/classification , Sarcoma, Small Cell/genetics , Sarcoma, Small Cell/pathology , Sarcoma, Small Cell/classification , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Diagnosis, Differential , Immunohistochemistry , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/classification , RNA-Binding Protein EWS/genetics , Repressor Proteins/genetics , Gene Rearrangement , Proto-Oncogene Proteins/genetics , Predictive Value of Tests , Phenotype , Genetic Predisposition to Disease , Oncogene Proteins, Fusion/genetics
2.
Diagn Pathol ; 18(1): 86, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37518334

ABSTRACT

ALES is a rare subtype that demonstrates the EWSR1-FLI1 translocation characteristic of ES and demonstrates complex epithelial differentiation including diffuse cytokeratin and p40 expression. It has predominantly recognized in the head and neck and is common in middle-aged population. This case is the first case of ALES reported in the pancreatic tail, sharing some morphological characteristics with ALES in the head and neck, including monotonous cytology, infiltrative growth pattern, and complex epithelioid differentiation, but ALES in the head and neck often has high-grade histological features (e.g., necrosis, high mitotic rate, etc.), and sudden keratinization can also occur, but these features were not reflected in this primary pancreatic tail ALES. Although ALES arising in the pancreatic tail and in the head and neck sites share the immunohistochemical and molecular profile, our case can provide new ideas in differential diagnosis of ALES arising in pancreatic tail and promote increased recognition and understanding of ALES.


Subject(s)
Adamantinoma , Neuroectodermal Tumors, Primitive, Peripheral , Sarcoma, Ewing , Skin Neoplasms , Middle Aged , Humans , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/genetics , Sarcoma, Ewing/chemistry , Adamantinoma/chemistry , Adamantinoma/pathology , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Skin Neoplasms/pathology , Cytodiagnosis
3.
Am J Surg Pathol ; 45(4): 523-530, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33538423

ABSTRACT

Ewing sarcoma (ES) is a highly malignant tumor that rarely occurs in the uterine cervix. Herein, we report 8 cases with ES arising primarily in the uterine cervix by focusing on clinicopathologic and molecular cytogenetic features and differential diagnoses. Eight cases of cervical ES were diagnosed between February, 2012, and September, 2018. The age of patients ranged from 13 to 47 years. Abnormal vaginal bleeding and lower abdominal pain were the most common symptoms. Histologically, the tumor was composed of uniform, round, and oval cells with a narrow rim of eosinophilic cytoplasm. Fibrous septa were observed between tumor cell nests. The tumors showed brisk mitotic activity and areas of coagulative necrosis. According to immunohistochemical studies, 50% (4/8) of the cases were positive for cytokeratin (AE1/AE3), and 87.5% (7/8) were positive for synaptophysin, which resulted in a diagnostic confusion with small cell carcinoma, primarily when dealing with small cervical biopsies. Molecular testing demonstrated the rearrangement of the EWSR1 gene in all of the 8 cases, which confirmed the diagnosis of ES. Although rare, ES should be considered as indicators of cervical small round cell neoplasms. Molecular analysis may greatly contribute to the final diagnosis of ES occurring in this unusual location.


Subject(s)
Biomarkers, Tumor , Sarcoma, Ewing , Uterine Neoplasms , Adolescent , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Diagnosis, Differential , Female , Gene Rearrangement , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Keratins/analysis , Middle Aged , Predictive Value of Tests , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Uterine Neoplasms/chemistry , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , Young Adult
4.
Int J Radiat Oncol Biol Phys ; 109(5): 1454-1471, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33373655

ABSTRACT

PURPOSE: The oncogenic EWS:Fli1 fusion protein is a key transcriptional mediator of Ewing sarcoma initiation, progression, and therapeutic resistance. Mithramycin A (MithA) is a potent and specific inhibitor of transcription mediated by the EWS:Fli1. We tested the hypothesis that pretreatment with MithA could selectively radiosensitize EWS:Fli1+ tumor cells by altering the transcriptional response to radiation injury. METHODS AND MATERIALS: A panel of 4 EWS:Fli1+ and 3 EWS:Fli1- Ewing sarcoma cell lines and 1 nontumor cell line were subjected to MithA dose-response viability assays to determine the relative potency of MithA in cells possessing or lacking the EWS:Fli1 fusion. Radiosensitization by MithA was evaluated by clonogenic survival assays in vitro and in a murine xenograft model. DNA damage was evaluated by comet assay and γ-H2Ax flow cytometry. Immunoblotting, flow cytometry, and reverse-transcription, polymerase chain reaction were used to evaluate DNA damage-induced signaling and repair processes and apoptosis. RESULTS: We found that MithA alone could potently and selectively inhibit the growth of EWS:Fli1+ tumor cells, but not cells lacking this fusion. Pretreatment with MithA for 24 hours before irradiation significantly reduced clonogenic survival in vitro and delayed tumor regrowth in vivo, prolonging survival of EWS:Fli1+ tumor-bearing mice. Although MithA did not increase the level of DNA double-strand breaks, mechanistic studies revealed that MithA pretreatment selectively inhibited DNA double-strand break repair through downregulation of EWS:Fli1-mediated transcription, leading to tumor cell death by apoptosis. CONCLUSIONS: Our data indicate that MithA is an effective radiosensitizer of EWS:Fli1+ tumors and may achieve better local control at lower doses of radiation.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/drug effects , Oncogene Proteins, Fusion/metabolism , Plicamycin/analogs & derivatives , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Radiation-Sensitizing Agents/pharmacology , Sarcoma, Ewing/radiotherapy , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , Dose-Response Relationship, Drug , Down-Regulation , Histones/metabolism , Mice , Plicamycin/pharmacology , Radiation Tolerance , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Xenograft Model Antitumor Assays
5.
Pathol Res Pract ; 215(12): 152665, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31585812

ABSTRACT

We describe the case of a Ewing sarcoma with prominent myxoid stroma of the temporal bone in a 26-year-old female. Histologically, the tumor exhibited a fascicular growth pattern of round to spindled cells with a minimal amount of pale eosinophilic to clear cytoplasm and oval or spindled nuclei with finely dispersed chromatin and small nucleoli. Myxoid changes were prominent (>50%), with reticular or pseudoacinar growth of the loosely cohesive cells. The tumor showed strong expression of CD99, FLI1, and CD56 and was positive for the EWSR1-FLI1 fusion transcript. The diagnosis of Ewing sarcoma with myxoid stroma (myxoid variant) is particularly problematic owing to the large number of potential mimics. The tumor extends the morphologic spectrum of Ewing sarcoma beyond the previously described histological variants, and broadens the differential diagnosis. For any round/spindle cell sarcoma, prominent myxoid stroma and CD99 immunoreactivity should prompt consideration for molecular studies that include analysis of both EWSR1 and FLI1.


Subject(s)
Bone Neoplasms/pathology , Sarcoma, Ewing/pathology , Stromal Cells/pathology , Temporal Bone/pathology , 12E7 Antigen/analysis , Adult , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Bone Neoplasms/chemistry , Bone Neoplasms/genetics , Bone Neoplasms/therapy , CD56 Antigen/analysis , Diagnosis, Differential , Female , Gene Fusion , Humans , Oncogene Proteins, Fusion/genetics , Predictive Value of Tests , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/therapy , Stromal Cells/chemistry , Temporal Bone/chemistry
6.
Rev Esp Patol ; 51(4): 239-243, 2018.
Article in Spanish | MEDLINE | ID: mdl-30269775

ABSTRACT

We report a case of Ewing sarcoma localized in the prostate gland of a 33-year-old patient without bone or soft tissue involvement. Evidence of EWS and FLI1 gene translocation was detected by fluorescence in situ hybridization (FISH). This is an unusual case with an interesting clinical presentation; indeed, only a few cases have been reported to date and not all have the supporting biological studies now considered essential for the diagnosis.


Subject(s)
Prostatic Neoplasms/pathology , Sarcoma, Ewing/pathology , Adult , Biomarkers, Tumor/analysis , Biopsy , Humans , In Situ Hybridization, Fluorescence , Male , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/genetics , Proto-Oncogene Protein c-fli-1/analysis , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS/analysis , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/diagnostic imaging , Sarcoma, Ewing/genetics
7.
Adv Anat Pathol ; 25(5): 314-326, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29911999

ABSTRACT

The diagnosis of small round cell tumors always has been extremely difficult, and our current classification systems continue to evolve. Since its initial discovery by Dr James Ewing, the historical context of what is acceptably included under the designation "Ewing sarcoma" has changed. Although Ewing sarcoma and primitive neuroectodermal tumor were both initially described in the early 20th century, these tumors were considered likely distinct entities until the end of that same century, almost 75 years later. With modern immunohistochemistry and more recent advances in molecular techniques, the understanding of Ewing sarcoma and Ewing-like tumors has improved dramatically but also raises new questions and challenges. We now know that this category of tumors is remarkably more heterogenous than initially thought, especially in regards to its cytogenetics and molecular properties, and some of these differences likely have prognostic relevance. Whether we are now expanding the spectrum of Ewing sarcoma or simply recognizing new entities is controversial. Therapeutic approaches to address these new categories and/or entities need further focus and attention. Herein, we provide a comprehensive historical perspective on Ewing sarcoma, Ewing-like tumors (CIC and BCOR-rearranged sarcomas), and related and/or similar small round cell tumors, often included in the differential diagnosis, including mesenchymal chondrosarcoma, desmoplastic small round cell tumor, and small cell osteosarcoma. We also seek to provide updates and insights into the evolving classification and clinical relevance of the Ewing family of tumors.


Subject(s)
Bone Neoplasms/pathology , Pathology , Sarcoma, Ewing/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Bone Neoplasms/chemistry , Bone Neoplasms/genetics , Bone Neoplasms/history , Diagnosis, Differential , Diffusion of Innovation , Forecasting , History, 20th Century , History, 21st Century , Humans , Immunohistochemistry , Molecular Diagnostic Techniques , Pathology/history , Pathology/trends , Predictive Value of Tests , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/history
8.
Am J Dermatopathol ; 40(9): 671-675, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29533277

ABSTRACT

Neuroendocrine differentiation or aberrant expression of neuroendocrine markers is very uncommon in angiosarcomas (AS) and creates a challenging differential diagnosis with other superficial or soft tissue tumors. Herein, we report a new case of superficial AS presenting as a tumor lesion on the little finger of the right hand of a 52-year-old man. The tumor displayed CD56, chromogranin-A, and synaptophysin immunoreactivity. Tumor cells were positive for vascular markers (CD31, FLI1, ERG, D2-40, VE-cadherin, VEGR1,2, and 3), CD99, and EMA, but were negative for S100, CK (AE1/AE3), CK20, polyomavirus, and myogenic (desmin and myogenin) and melanocyte markers (melan-A and HMB45). Ki67 immunostains indicated high proliferative activity (>50%). The whole-body computed tomography did not reveal distant disease. The initial assessment considered several tumor subtypes as possible histological diagnoses, including Ewing sarcoma, Ewing-like sarcoma, Merkel cell carcinoma, and undifferentiated "small round cell sarcoma". Fluorescence in situ hybridization analysis was negative for EWSR1 translocation and molecular analysis failed to detect any EWSR1, CIC, SYT or BCOR rearrangement. As a follow-up investigation, we tested 17 cutaneous/superficial AS for neuroendocrine markers; however, only one of these showed focal CD56 and synaptophysin expression. In conclusion, the present findings indicate that neuroendocrine differentiation is a very infrequent feature in AS. We report an AS of the finger with an uncommon histological appearance and immunohistochemical profile: predominant round cell tumor proliferation and neuroendocrine differentiation. Pathologists should be aware of these potential histological and immunohistochemical pitfalls in AS.


Subject(s)
Carcinoma, Neuroendocrine/pathology , Cell Differentiation , Hemangiosarcoma/pathology , Sarcoma, Ewing/pathology , Sarcoma, Small Cell/pathology , Skin Neoplasms/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Carcinoma, Neuroendocrine/chemistry , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/surgery , Cell Proliferation , Diagnosis, Differential , Fingers , Hemangiosarcoma/chemistry , Hemangiosarcoma/genetics , Hemangiosarcoma/surgery , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Predictive Value of Tests , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Small Cell/chemistry , Sarcoma, Small Cell/genetics , Sarcoma, Small Cell/surgery , Skin Neoplasms/chemistry , Skin Neoplasms/genetics , Skin Neoplasms/surgery
9.
Zhonghua Bing Li Xue Za Zhi ; 46(11): 750-755, 2017 Nov 08.
Article in Chinese | MEDLINE | ID: mdl-29136686

ABSTRACT

Objective: To investigate the clinicopathologic and molecular characteristics, diagnostic, differential diagnostic and prognostic features of malignant gastrointestinal neuroectodermal tumor. Methods: Two cases of malignant gastrointestinal neuroectodermal tumor were retrieved; the clinical and radiologic features, histomorphology, immunophenotype, molecular genetics and prognosis were analyzed and the relevant literature reviewed. Results: Case 1 was a 57-year-old male, presented with recurrent abdominal pain and melena. Pelvic imaging showed a circumscribed thickening of the wall of a small intestinal segment, and a malignant lymphoma was favored. Case 2 was a 24-year-old male, presented with recurrent small intestinal malignancy. Imaging demonstrated multiple masses in the peritoneal and pelvic cavities, and a malignant gastrointestinal stromal tumor with multiple metastases was suspected. Grossly both tumors were located mainly in the muscularis propria of small intestine. Case 1 showed a single 5.5 cm tumor; and case 2 consisted of two tumors measuring 4 cm and 6 cm respectively. Microscopic examination of both tumors showed small round blue, but focally spindled or clear tumor cells in solid pattern. The tumor cells had scanty cytoplasm, indistinctive nucleoli and brisk mitoses. Osteoclast-like giant cells were dispersed within the stroma. In case 1 rosette-like and pseudo-papillary growth patterns were noted, and in case 2 there were variable-sized hemorrhagic cysts. By immunohistochemistry, both tumors showed strong and diffuse expression of SOX10 and S-100, and focal to diffuse expression of neuroendocrine markers (CD56 or synaptophysin). Case 2 exhibited focal reactivity to pan-cytokeratin. Both tumors lacked expression of markers associated with gastrointestinal stromal tumor, smooth muscle tumor, melanoma (HMB45 or Melan A), dendritic cell tumor and Ewing sarcoma. Fluorescence in situ hybridization analysis demonstrated EWSR1 rearrangement in both tumors and the next generation sequencing confirmed EWSR1-ATF1 gene fusion in case 2. At follow-up of 16 months, case 1 was recurrence or metastasis free; whereas case 2 showed multiple recurrences and metastases within 19 months although stable disease was transiently achieved when treated with combinations of multidrug and targeted chemotherapy. Conclusions: Malignant gastrointestinal neuroectodermal tumor is a rare and aggressive soft tissue sarcoma with a predilection for small intestine. It has distinctive morphologic, immunohistochemical and molecular characteristics and needs to be distinguished from other small blue round and spindle cell tumors that occur in the gut. Careful attentions to its characteristic histomorphology with the judicious use of immunohistochemistry and molecular genetics can help to distinguish this tumor from its many mimickers.


Subject(s)
Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Neuroectodermal Tumors/pathology , Adult , Biomarkers, Tumor/analysis , Calmodulin-Binding Proteins/analysis , Diagnosis, Differential , Gastrointestinal Neoplasms/chemistry , Gastrointestinal Stromal Tumors/chemistry , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Keratins/analysis , MART-1 Antigen , Male , Melanoma/chemistry , Melanoma/pathology , Middle Aged , Neoplasm Recurrence, Local , Neuroectodermal Tumors/chemistry , RNA-Binding Proteins , S100 Proteins/analysis , SOXE Transcription Factors/analysis , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/pathology , Synaptophysin/analysis
10.
Cancer ; 123(19): 3807-3815, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28608950

ABSTRACT

BACKGROUND: Programmed death 1 (PD-1) signaling in the tumor microenvironment dampens immune responses to cancer, and blocking this axis induces antitumor effects in several malignancies. Clinical studies of PD-1 blockade are only now being initiated in pediatric patients, and little is known regarding programmed death-ligand 1 (PD-L1) expression in common childhood cancers. The authors characterized PD-L1 expression and tumor-associated immune cells (TAICs) (lymphocytes and macrophages) in common pediatric cancers. METHODS: Whole slide sections and tissue microarrays were evaluated by immunohistochemistry for PD-L1 expression and for the presence of TAICs. TAICs were also screened for PD-L1 expression. RESULTS: Thirty-nine of 451 evaluable tumors (9%) expressed PD-L1 in at least 1% of tumor cells. The highest frequency histotypes comprised Burkitt lymphoma (80%; 8 of 10 tumors), glioblastoma multiforme (36%; 5 of 14 tumors), and neuroblastoma (14%; 17 of 118 tumors). PD-L1 staining was associated with inferior survival among patients with neuroblastoma (P = .004). Seventy-four percent of tumors contained lymphocytes and/or macrophages. Macrophages were significantly more likely to be identified in PD-L1-positive versus PD-L1-negative tumors (P < .001). CONCLUSIONS: A subset of diagnostic pediatric cancers exhibit PD-L1 expression, whereas a much larger fraction demonstrates infiltration with tumor-associated lymphocytes. PD-L1 expression may be a biomarker for poor outcome in neuroblastoma. Further preclinical and clinical investigation will define the predictive nature of PD-L1 expression in childhood cancers both at diagnosis and after exposure to chemoradiotherapy. Cancer 2017;123:3807-3815. © 2017 American Cancer Society.


Subject(s)
B7-H1 Antigen/analysis , Lymphocytes, Tumor-Infiltrating , Macrophages , Neoplasm Proteins/analysis , Neoplasms/chemistry , Bone Neoplasms/chemistry , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Burkitt Lymphoma/chemistry , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Child , Glioblastoma/chemistry , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Immunohistochemistry , Neoplasms/immunology , Neoplasms/mortality , Neoplasms/pathology , Neuroblastoma/chemistry , Neuroblastoma/immunology , Neuroblastoma/mortality , Neuroblastoma/pathology , Osteosarcoma/chemistry , Osteosarcoma/immunology , Osteosarcoma/pathology , Rhabdomyosarcoma/chemistry , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/immunology , Sarcoma, Ewing/pathology , Tissue Array Analysis
11.
Diagn Pathol ; 12(1): 37, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28472972

ABSTRACT

BACKGROUND: Ewing's sarcoma (ES) and primitive neuroectodermal tumors (PNET) are closely related tumors. Although soft tissue ES/PNET are common in clinical practice, they are rare in the small intestine. Because of the absence of characteristic clinical symptoms, they are easily misdiagnosed as other benign or malignant diseases. CASE PRESENTATION: Here, we present the case of a 16-year-old female who complained of anemia and interval hematochezia. Her serum test results showed only a slight elevation of CA-125 and a low level of hemoglobin. Computer tomography and magnetic resonance imaging revealed a cystic and solid mass in the lower abdominal quadrant and pelvic region, which prompted suspicion of a malignant gastrointestinal stromal tumor of the small intestine. After resection, the tumor's histology and immunohistochemistry (positive for CD99, vimentin and synaptophysin) results suggested ES/PNET. Fluorescent in situ hybridization tests proved the breakpoint rearrangement of the EWSR1 gene in chr 22.Ultrastructural analysis revealed neurosecretory and glycogen granules in the tumor cell cytoplasm. CONCLUSIONS: Together, these data supported the diagnosis of a rare case of localized ES/PNET in the small intestine without adjuvant chemo- or radiotherapy. To our knowledge, this is the first report from China of a primary small bowel ES/PNET in the English-language literature. In addition, on the basis of findings from previous publications and the current case, the optimal treatment for localized gastrointestinal ES/PNET is discussed.


Subject(s)
Ileal Neoplasms/pathology , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Sarcoma, Ewing/pathology , Adolescent , Biomarkers, Tumor/genetics , Biopsy , China , Female , Gene Rearrangement , Humans , Ileal Neoplasms/chemistry , Ileal Neoplasms/genetics , Ileal Neoplasms/surgery , Immunohistochemistry , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Neuroectodermal Tumors, Primitive, Peripheral/chemistry , Neuroectodermal Tumors, Primitive, Peripheral/genetics , Neuroectodermal Tumors, Primitive, Peripheral/surgery , RNA-Binding Protein EWS/genetics , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/surgery , Tomography, X-Ray Computed
12.
Zhonghua Bing Li Xue Za Zhi ; 46(2): 102-107, 2017 Feb 08.
Article in Chinese | MEDLINE | ID: mdl-28173669

ABSTRACT

Objective: To investigate the morphologic, immunohistochemical, genetic, clinical features and prognosis of Ewing-like BCOR-CCNB3 gene fusion undifferentiated sarcoma (BCOR-CCNB3 fusion sarcoma). Method: Seventeen Ewing-like sarcoma cases were screened for CCNB3 expression and BCOR-CCNB3 fusion transcripts by immunohistochemistry and RT-PCR among 260 cases of Ewing-like sarcomas collected during Jan, 2006 to Dec, 2015. Three cases of BCOR-CCNB3 fusion sarcoma were found among 17 atypical Ewing sarcomas, and follow-up were conducted. Results: The harboring of BCOR-CCNB3 fusion transcript was confirmed by RT-PCR and directly sequencing results. The three patients aged between 8 and 11 years old. Two of them were male and the other one was female. One patient achieved a complete response after chemotherapy, the other two died without chemotherapy after surgical excision in 12 months. Tumor cells in all 3 cases showed diffuse nuclear CCNB3, TLE1 and cyclin D1 positivity, while CCNB3 (0/12), TLE1 (1/12) and cyclin D1 (4/12) positivity was infrequent in the 12 cases of classical Ewing's sarcoma. The oval or plump spindle tumor cells with fine chromatin arranged in solid pattern, the nucleoli was inconspicuous. The delicate capillary networks were obvious in the tumor. Conclusion: With a detailed description of the histological spectrum, immunohistochemical features and clinical characteristic of BCOR-CCNB3 sarcoma, justify distinction from Ewing sarcoma could be possible.


Subject(s)
Sarcoma, Ewing , Sarcoma , Child , Cyclin B/metabolism , Cyclin D1/metabolism , Diagnosis, Differential , Fatal Outcome , Female , Humans , Immunohistochemistry , Male , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion , Prognosis , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sarcoma/chemistry , Sarcoma/genetics , Sarcoma/mortality , Sarcoma/pathology , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology
13.
Hum Pathol ; 60: 58-65, 2017 02.
Article in English | MEDLINE | ID: mdl-27984122

ABSTRACT

Small round blue cell tumors (SRBCTs) of children and adolescents are often diagnostically challenging lesions. With the increasing diagnostic approach based on small biopsies, there is the need of specific immunomarkers that can help in the differential diagnosis among the different tumor histotypes to assure the patient a correct diagnosis for proper treatment. Based on our recent studies showing cyclin D1 overexpression in both Ewing sarcoma/primitive peripheral neuroectodermal tumor (EWS/pPNET) and peripheral neuroblastic tumors (neuroblastoma and ganglioneuroblastoma), we immunohistochemically assessed cyclin D1 immunoreactivity in 128 cases of SRBCTs in children and adolescents to establish its potential utility in the differential diagnosis. All cases of EWS/pPNET and the undifferentiated/poorly differentiated neuroblastomatous component of all peripheral neuroblastic tumors exhibited strong and diffuse nuclear staining (>50% of neoplastic cells) for cyclin D1. In contrast, this marker was absent from rhabdomyosarcoma (regardless of subtype) and lymphoblastic lymphoma (either B- or T-cell precursors), whereas it was only focally detected (<5% of neoplastic cells) in some cases of Wilms tumor (blastemal component) and desmoplastic small round cell tumor. Our findings suggest that cyclin D1 can be exploitable as a diagnostic adjunct to conventional markers in confirming the diagnosis of EWS/pPNET or neuroblastoma/ganglioneuroblastoma. Its use in routine practice may also be helpful for those cases of SRBCT with undifferentiated morphology that are difficult to diagnose after application of the conventional markers.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/chemistry , Cyclin D1/analysis , Ganglioneuroblastoma/chemistry , Neuroblastoma/chemistry , Neuroectodermal Tumors, Primitive, Peripheral/chemistry , Sarcoma, Ewing/chemistry , Adolescent , Biopsy , Bone Neoplasms/pathology , Cell Differentiation , Child , Child, Preschool , Desmoplastic Small Round Cell Tumor/chemistry , Desmoplastic Small Round Cell Tumor/pathology , Diagnosis, Differential , Female , Ganglioneuroblastoma/pathology , Humans , Immunohistochemistry , Infant , Kidney Neoplasms/chemistry , Kidney Neoplasms/pathology , Male , Neuroblastoma/pathology , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Predictive Value of Tests , Retrospective Studies , Rhabdomyosarcoma/chemistry , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/pathology , Wilms Tumor/chemistry , Wilms Tumor/pathology , Young Adult
14.
Hum Pathol ; 55: 91-100, 2016 09.
Article in English | MEDLINE | ID: mdl-27246176

ABSTRACT

Sarcomas have traditionally been classified according to their chromosomal alterations regardless of whether they accompany simple or complex genetic changes. Ewing sarcoma, a classic small round cell bone tumor, is a well-known mesenchymal malignancy that results from simple sarcoma-specific genetic alterations. The genetic alterations are translocations between genes of the TET/FET family (TLS/FUS, EWSR1, and TAF15) and genes of the E26 transformation-specific (ETS) family. In this review, we intend to summarize a chronicle of molecular findings of Ewing sarcoma including recent advances and explain resultant molecular pathogenesis.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Sarcoma, Ewing/genetics , 12E7 Antigen/analysis , Biomarkers, Tumor/analysis , Bone Neoplasms/chemistry , Bone Neoplasms/pathology , Chromosome Aberrations , Diagnosis, Differential , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Molecular Diagnostic Techniques , Phenotype , Predictive Value of Tests , Prognosis , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/pathology , Transcription, Genetic , Translocation, Genetic
15.
Am J Surg Pathol ; 40(3): 313-23, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26685084

ABSTRACT

The CIC gene rearrangement exists in a subset of small round cell sarcomas. As the nosologic relationship of these sarcomas to Ewing sarcomas remains undetermined, we examined 20 CIC-rearranged sarcomas to compare their clinicopathologic features with those of Ewing sarcomas. The CIC-rearranged sarcomas were from a group of 14 men and 6 women with a median age of 24.5 years. The primary tumor sites included the limbs, trunk wall, internal trunk, lung, cerebrum, and pharynx. A comparison of the demographic and clinical characteristics of the 20 patients with CIC-rearranged sarcomas with those of the 53 near-consecutive patients with EWSR1-rarranged Ewing sarcomas showed that there were no differences with respect to their ages and sexes. Although none of the CIC-rearranged sarcomas arose in the bone, 40% of the Ewing sarcomas primarily affected the skeleton. The overall survival of patients with Ewing sarcomas was significantly better than that for patients with CIC-rearranged sarcomas. A histologic comparison of the CIC-rearranged sarcomas with 20 EWSR1-rearranged Ewing sarcomas showed significantly higher degrees of lobulation, nuclear pleomorphism, the prominence of the nucleoli, spindle cell elements, and myxoid changes in the CIC-rearranged sarcomas. Distinguishing immunohistochemical features included heterogenous CD99 reactivity, nuclear WT1 expression, and calretinin expression in the CIC-rearranged sarcomas and NKX2.2 expression in the Ewing sarcomas. CIC-rearranged sarcomas are distinct from Ewing sarcomas clinically, morphologically, and immunohistochemically, and they should be considered a separate entity rather than being grouped within the same family of tumors.


Subject(s)
Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Calmodulin-Binding Proteins/genetics , Gene Rearrangement , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Sarcoma, Ewing/genetics , Sarcoma/genetics , Adolescent , Adult , Aged , Bone Neoplasms/chemistry , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Diagnosis, Differential , Female , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Male , Middle Aged , Nuclear Proteins , Predictive Value of Tests , Prognosis , RNA-Binding Protein EWS , Sarcoma/chemistry , Sarcoma/mortality , Sarcoma/pathology , Sarcoma/therapy , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/mortality , Sarcoma, Ewing/pathology , Sarcoma, Ewing/therapy , Transcription Factors , Young Adult
16.
Int J Clin Exp Pathol ; 8(6): 7575-82, 2015.
Article in English | MEDLINE | ID: mdl-26261672

ABSTRACT

Ewing's sarcoma is the second most common pediatric bone tumor. Primary Ewing's sarcoma occurring in the cerebral cranium is exceptionally rare, with only one reported case of multiple tumor lesions in adolescence to date. We report a case of a 5-year-old male patient with multiple primary Ewing's sarcomas associated with the cranial bones, the first pediatric case report to date. We also review 71 cases Ewing's sarcoma involving intracranial extension. The purpose of this article is to provide data concerning the clinical and therapeutic course of multiple primary Ewing's sarcomas in associated with cerebral cranium.


Subject(s)
Neoplasms, Multiple Primary/pathology , Sarcoma, Ewing/pathology , Skull Neoplasms/pathology , Biomarkers, Tumor/analysis , Child, Preschool , Craniotomy , Disease Progression , Fatal Outcome , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Neoplasms, Multiple Primary/chemistry , Neoplasms, Multiple Primary/surgery , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/surgery , Skull Neoplasms/chemistry , Skull Neoplasms/surgery , Time Factors , Tomography, X-Ray Computed , Treatment Outcome
17.
Am J Surg Pathol ; 39(9): 1197-205, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26135561

ABSTRACT

Ovarian small cell carcinoma of the hypercalcemic type (SCCOHT)/ovarian rhabdoid tumor is a rare and highly malignant tumor that typically occurs in young women. Up until now the diagnosis has been made on the basis of morphology without any specific immunohistochemical (IHC) markers. However, several authors have shown recently that SCCOHTs are characterized by inactivation of the SMARCA4 gene (encoding the BRG1 protein) resulting in a loss of BRG1 protein expression in IHC. We evaluated BRG1 and INI1 expression in 12 SCCOHTs and in a series of 122 tumors that could mimic SCCOHT morphologically: 9 juvenile granulosa cell tumors, 47 adult granulosa cell tumors, 33 high-grade ovarian serous carcinomas, 9 desmoplastic round cell tumors, 13 Ewing sarcomas (5 from the pelvis and 8 from soft tissues), 1 round cell sarcoma associated with CIC-DUX4 translocation from soft tissue (thigh), 1 case of high-grade endometrial stromal sarcoma of the ovary, and 9 melanomas. Forty-four adult granulosa cell tumors were interpretable by IHC. All 12 SCCOHTs were devoid of BRG1 expression and expressed INI1. All other interpretable 119 tumors showed BRG1 nuclear positivity, with variable staining proportions, ranging from 10% to 100% of positive cells (mean: 77%, median: 80%), variable intensities (weak: 5%, moderate: 37%, strong: 58%), and distributions: diffuse in 82 cases (70%) and heterogenous in 36 cases (30%). BRG1 positivity was heterogenous in desmoplastic round cell tumors and adult granulosa cell tumors. Overall, BRG1 is a useful diagnostic marker in SCCOHT, showing the absence of expression in SCCOHT. Nevertheless, the possible heterogeneity and the variable intensity of this staining warrant caution in the interpretation of BRG1 staining in biopsy specimens.


Subject(s)
Biomarkers, Tumor/analysis , Bone Neoplasms/chemistry , Carcinoma, Small Cell/chemistry , DNA Helicases/analysis , Hypercalcemia/metabolism , Melanoma/chemistry , Nuclear Proteins/analysis , Ovarian Neoplasms/chemistry , Rhabdoid Tumor/chemistry , Sarcoma, Ewing/chemistry , Soft Tissue Neoplasms/chemistry , Transcription Factors/analysis , Adolescent , Adult , Biomarkers, Tumor/genetics , Biopsy , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Diagnosis, Differential , Endometrial Neoplasms/chemistry , Endometrial Neoplasms/pathology , Female , Granulosa Cell Tumor/chemistry , Granulosa Cell Tumor/pathology , Humans , Hypercalcemia/genetics , Hypercalcemia/pathology , Immunohistochemistry , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Predictive Value of Tests , Rhabdoid Tumor/genetics , Rhabdoid Tumor/pathology , Sarcoma, Endometrial Stromal/chemistry , Sarcoma, Endometrial Stromal/pathology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Young Adult
18.
Pathol Res Pract ; 211(9): 688-92, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26163185

ABSTRACT

Very recently a new designation of "Malignant Neuroectodermal Gastrointestinal Tumor" has been proposed for an aggressive form of neuroectodermal tumor with features similar to that of Clear Cell Sarcoma of Soft Tissue, however without a melanocytic differentiation. Also known as "clear cell sarcoma-like tumors of the gastrointestinal tract", these tumors show some features strongly suggesting an origin from a gastrointestinal neuroectodermal precursor cell unable to differentiate along the melanocytic lineage. They occur mainly in young and middle-aged adults, and have a poor prognosis with a high rate of liver and lymphnode metastases. Histologically they are composed of epithelioid or oval-to spindle cells with a sheet-like or nested pattern of growth, strongly positive for neural markers (S-100, SOX10, and vimentin) and negative for the melanocytic ones. EWSR1 gene rearrangements including EWSR1-ATF1 or EWSR1-CREB1 GENE fusions are typically assessed in these tumors. Here we report a case of malignant neuroectodermal gastrointestinal tumor which immunophenotypically unusually expressed FLI-1, occurring in a 29-year-old man with a previous medical history of Ewing sarcoma. We finally suggest that this case might be a further evidence of a link between these two entities.


Subject(s)
Bone Neoplasms/pathology , Neoplasms, Second Primary/pathology , Neuroectodermal Tumors, Primitive/pathology , Sarcoma, Clear Cell/pathology , Sarcoma, Ewing/pathology , Stomach Neoplasms/pathology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Biopsy , Bone Neoplasms/chemistry , Chemotherapy, Adjuvant , Gastrectomy , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Magnetic Resonance Imaging , Male , Neoplasms, Second Primary/chemistry , Neoplasms, Second Primary/genetics , Neoplasms, Second Primary/therapy , Neuroectodermal Tumors, Primitive/chemistry , Neuroectodermal Tumors, Primitive/genetics , Neuroectodermal Tumors, Primitive/therapy , Polymerase Chain Reaction , Sarcoma, Clear Cell/chemistry , Sarcoma, Clear Cell/genetics , Sarcoma, Clear Cell/therapy , Sarcoma, Ewing/chemistry , Stomach Neoplasms/chemistry , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Treatment Outcome
19.
Am J Surg Pathol ; 39(9): 1267-74, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26034869

ABSTRACT

Ewing sarcoma family tumors (EFTs) of the head and neck are rare and may be difficult to diagnose, as they display significant histologic overlap with other more common undifferentiated small blue round cell malignancies. Occasionally, EFTs may exhibit overt epithelial differentiation in the form of diffuse cytokeratin immunoexpression or squamous pearls, resembling the so-called adamantinoma-like EFTs and being challenging to distinguish from bona fide carcinomas. Furthermore, the presence of EWSR1 gene rearrangement correlated with strong keratin expression may suggest a myoepithelial carcinoma. Herein, we analyze a series of 7 adamantinoma-like EFTs of the head and neck, most of them being initially misdiagnosed as carcinomas because of their anatomic location and strong cytokeratin immunoexpression, and subsequently reclassified as EFT by molecular techniques. The tumors arose in the sinonasal tract (n=2), parotid gland (n=2), thyroid gland (n=2), and orbit (n=1), in patients ranging in age from 7 to 56 years (mean, 31 y). Microscopically, they departed from the typical EFT morphology by growing as nests with peripheral nuclear palisading and prominent interlobular fibrosis, imparting a distinctly basaloid appearance. Moreover, 2 cases exhibited overt keratinization in the form of squamous pearls, and 1 sinonasal tumor demonstrated areas of intraepithelial growth. All cases were positive for CD99, pancytokeratin, and p40. A subset of cases showed synaptophysin, S100 protein, and/or p16 reactivity, further confounding the diagnosis. Fluorescence in situ hybridization assays showed EWSR1 and FLI1 rearrangements in all cases. Our results reinforce that a subset of head and neck EFTs may show strong cytokeratin expression or focal keratinization, and are therefore histologically indistinguishable from more common true epithelial neoplasms. Thus, CD99 should be included in the immunopanel of a round cell malignancy regardless of strong cytokeratin expression or anatomic location, and a strong and diffuse CD99 positivity should prompt molecular testing for the presence of EWSR1 gene rearrangements.


Subject(s)
Adamantinoma/diagnosis , Bone Neoplasms/diagnosis , Head and Neck Neoplasms/diagnosis , Myoepithelioma/diagnosis , Neuroectodermal Tumors, Primitive, Peripheral/diagnosis , Sarcoma, Ewing/diagnosis , 12E7 Antigen , Adamantinoma/chemistry , Adamantinoma/genetics , Adamantinoma/pathology , Adolescent , Adult , Antigens, CD/analysis , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Bone Neoplasms/chemistry , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Calmodulin-Binding Proteins/genetics , Cell Adhesion Molecules/analysis , Cell Differentiation , Child , Diagnosis, Differential , Female , Gene Rearrangement , Head and Neck Neoplasms/chemistry , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Keratins/analysis , Male , Middle Aged , Myoepithelioma/chemistry , Myoepithelioma/genetics , Myoepithelioma/pathology , Neuroectodermal Tumors, Primitive, Peripheral/chemistry , Neuroectodermal Tumors, Primitive, Peripheral/genetics , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Predictive Value of Tests , Proto-Oncogene Protein c-fli-1/genetics , RNA-Binding Protein EWS , RNA-Binding Proteins/genetics , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Tissue Array Analysis , Young Adult
20.
Ann Pathol ; 35(1): 86-97, 2015 Jan.
Article in French | MEDLINE | ID: mdl-25534668

ABSTRACT

Ewing sarcoma family of tumors are mainly aggressive sarcomas of bone and also arising in soft tissues, which share common features: morphological features of basophilic round cell tumors, immunohistochemical features by expression of membrane CD99 protein, and genetic features with a translocation involving EWS and FLI1 in approximately 90% of cases. The discovery of this translocation has made it possible to unify in a single entity several lesions such as PNET, neuropitheliomas, Askin tumors, Ewing sarcomas… Since then, the extensive use of molecular/genetic methods has helped to identify an increasing number of molecular anomalies in unclassified round cell sarcomas, these sarcomas often harboring an atypical morphology and a less frequent CD99 positivity. Besides the rearrangements between the FET family of genes (EWS or FUS) and the wide ETS family of genes (FLI1, ERG, FEV, ETV…), new partner genes are gradually identified: cases with EWS-non ETS partners are extremely rare, but there are more important groups which are CIC-DUX4 and BCOR-CCNB3 translocation-positive sarcomas. These findings raise the problem of the nosological borders of the Ewing/PNET entity and its links with new "Ewing-like" groups of tumors, and raise the therapeutic problems. The forward-looking identification of new round cell sarcomas should enable studies of wider series to try to answer these questions.


Subject(s)
Bone Neoplasms/pathology , Neuroectodermal Tumors, Primitive/pathology , Sarcoma, Ewing/pathology , Soft Tissue Neoplasms/pathology , Biomarkers, Tumor , Bone Neoplasms/chemistry , Bone Neoplasms/classification , Bone Neoplasms/diagnosis , Bone Neoplasms/epidemiology , Bone Neoplasms/genetics , Diagnosis, Differential , Disease Progression , Humans , Neoplasm Invasiveness , Neuroectodermal Tumors, Primitive/chemistry , Neuroectodermal Tumors, Primitive/classification , Neuroectodermal Tumors, Primitive/diagnosis , Neuroectodermal Tumors, Primitive/epidemiology , Neuroectodermal Tumors, Primitive/genetics , Oncogene Proteins, Fusion/genetics , Sarcoma, Ewing/chemistry , Sarcoma, Ewing/classification , Sarcoma, Ewing/diagnosis , Sarcoma, Ewing/epidemiology , Sarcoma, Ewing/genetics , Soft Tissue Neoplasms/chemistry , Soft Tissue Neoplasms/classification , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/epidemiology , Soft Tissue Neoplasms/genetics , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...