Next Article in Journal
Impact of Intraoperative Nefopam on Postoperative Pain, Opioid Use, and Recovery Quality with Parietal Pain Block in Single-Port Robotic Cholecystectomy: A Prospective Randomized Controlled Trial
Next Article in Special Issue
Multiorgan Toxicity from Dual Checkpoint Inhibitor Therapy, Resulting in a Complete Response—A Case Report
Previous Article in Journal
Aspects of Histopathological and Ultrastructural Retinal Changes in Chronic Exposure to Hydroxychloroquine
Previous Article in Special Issue
Assessing the Therapeutic Impacts of HAMLET and FOLFOX on BRAF-Mutated Colorectal Cancer: A Study of Cancer Cell Survival and Mitochondrial Dynamics In Vitro and Ex Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Case Report

Intestinal Clear Cell Sarcoma—A Case Presentation of an Extremely Rare Tumor and Literature Review

1
General Surgery Department 10, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
3
Pathology Department, Bucharest Institute of Oncology “Prof. Dr. Al. Trestioreanu”, 022328 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
Medicina 2024, 60(6), 847; https://doi.org/10.3390/medicina60060847
Submission received: 29 April 2024 / Revised: 16 May 2024 / Accepted: 20 May 2024 / Published: 22 May 2024
(This article belongs to the Special Issue Development of Gastrointestinal Cancer Surgery)

Abstract

:
Background: Clear cell sarcoma (CCS) is an extremely rare form of sarcoma representing less than 1% of all soft-tissue sarcomas. It has morphological, structural, and immunohistochemical similarities to malignant melanoma, affecting young adults and equally affecting both sexes, and is usually located in the tendinous sheaths and aponeuroses of the limbs. Gastrointestinal localization is exceptional, with less than 100 cases reported thus far. The gene fusion of activating transcription factor 1 (ATF1) and the Ewing sarcoma breakpoint region 1 (EWSR1) are pathognomonic for clear cell sarcoma, representing the key to the diagnosis. CCS is an extremely aggressive tumor, with >30% having distant or lymphatic metastasis at the time of diagnostic, and it has a high recurrence rate of over 80% in the first year after diagnosis and a high tendency for metastatic dissemination. Given the rarity of this tumor, there is no standardized treatment. Early diagnosis and radical surgery are essential in the treatment of CCS both for the primary tumor and for recurrence or metastasis. Chemo-radiotherapy has very little effect and is rarely indicated, and the role of targeted therapies is still under investigation. Case presentation: We present an extremely rare case of intestinal CSS in a 44-year-old Caucasian female. The patient, asymptomatic, first presented for a routine checkup and was diagnosed with mild iron-deficiency anemia. Given her family history of multiple digestive cancers, additional investigations were requested (gastroscopy, colonoscopy, tumoral markers and imaging) and the results were all within normal limits. In the subsequent period, the patient experienced mild diffuse recurrent abdominal pain, which occurred every 2–3 months. Two years later, the patient presented with symptoms of intestinal obstruction and underwent an emergency laparotomy followed by segmental enterectomy and regional lymphadenectomy for stenotic tumor of the jejunum. Histology, immunohistochemistry, and genetic testing established the diagnosis of CCS. No adjuvant therapy was indicated. Initially, no signs of recurrence or metastasis were detected, but after 30 and 46 months, respectively, from the primary treatment, the patient developed liver metastasis and pericolic peritoneal implants treated by atypical hepatic resections and right hemicolectomy. The patient remains under observation.

1. Introduction

Clear cell sarcoma (CCS), also called “soft-tissue melanoma”, is an extremely rare malignant tumor and was first described by F. Enzinger in 1965. It poses diagnostic challenges due to similarities in structure, morphology and immunohistochemistry very much resembling those of malignant melanoma (MM) [1,2]. Among the common phenotypic characteristics shared with malignant melanoma are the presence of melanin, as well as the expression of melanoma-associated markers such as HMB-45, microphthalmia transcription factor (MiTF), S100 protein, and Melan-A [1]. From histological and immunohistochemical points of view, CCS and MM are almost identical, the two being differentiated by fluorescence in situ hybridization (FISH) and real-time Polymerase Chain Reaction (RT-PCR). The main key in the differential diagnosis is a reciprocal chromosomal translocation t(12;22) (q13;q12) leading to the occurrence of a fusion between two genes, namely the activating transcription factor 1 (ATF1) gene and the Ewing sarcoma breakpoint region 1 (EWSR1) gene, resulting in a fusion protein, EWSR1-ATF1. The presence of this gene fusion is a relevant indicator, as it occurs in the vast majority of patients. A variant fusion protein EWSR1-CREB1 can occur in gastrointestinal CCS resulting from the chromosomal translocation t(12;22) (q34;q12). Also, MM frequently presents BRAF mutations, which are absent in CCS.
CCS originates from the neural crest cells (as proven by the presence of melanosomes in the cytoplasm of tumoral cells), and represents less than 1% of all soft-tissue sarcomas, affecting young adults and equally affecting both sexes, and is usually located in the tendinous sheaths and aponeuroses of the limbs. In addition to MM, other differential diagnoses are made with Kaposi’s sarcoma and malignant peripheral nerve sheath tumor (MPNST). Kaposi’s sarcoma typically occurs in immunocompromised patients, while MPNST is often found in patients with neurofibromatosis type 1 [3].
The most common clinical presentation is a painful (pain present in 33–55% of cases [4]), rapidly growing tumoral mass located around the ankles. More than 90% of all CCS are in the extremities and the neck. There are also rare reports of CCS located inside the thoracic and abdominal cavity. Only 6–7% of CCS cases originate from the gastro-intestinal tract, making it an extremely rare form. To the best of our ability, we could only locate fewer than 100 cases in the international literature, most being solitary tumors and presenting as intestinal occlusion or anemic syndrome. Such tumors usually present with various nonspecific digestive symptoms (such as diffuse pain, colic, nausea, or vomiting) or general signs (fatigue, weight loss). Palpable abdominal masses are also possible.
Differential diagnostics of CCS with gastro-intestinal localization include MM, malignant gastrointestinal neuroectodermal tumors or GNET (which also present EWSR1-ATF1 or EWSR1-CREB1 fusion and S100 positivity, but lack melanocytic markers and frequently have gigantic osteoclast-like cells), clear-cell carcinomas (but these are cytokeratin-positive), gastrointestinal neuroendocrine tumors (differentiated by serologic test and scintigraphy), intestinal adenocarcinomas, and gastrointestinal stromal tumors (GIST).
Given the highly heterogeneous clinical presentation, clinicians, especially those without experience, can easily be misled, struggling to differentiate clear cell sarcoma from malignant melanoma correctly, especially for the intraabdominal localization. This, combined with the fact that immunohistochemistry cannot distinguish between the two types of tumors, means that clear cell sarcoma is misdiagnosed as malignant melanoma. Due to CCS rarity, FISH and RT-PCR testing are essential for an accurate diagnosis [3]. Correct and rapid diagnostics are necessary for insuring the optimal therapeutic response and a better oncological outcome, and physicians should always consider genetic testing for CCS when faced with an MM diagnostic.
CCS is an extremely aggressive tumor, with >30% of cases having distant or lymphatic metastasis at the time of initial diagnostic and a recurrence rate of over 80% in the first year after primary treatment. More than 60% of all cases will develop metastasis within 12 months from diagnostics. Several risk factors have been described for clear cell sarcoma, in the absence of a clear cause, including chemotherapy, radiotherapy, and genetic predisposition [4,5]. In the literature, we could only find a few studies focusing on prognostic factors and survival after CCS [6,7,8]. The largest of those studies, including 489 patients, determined that 38% of patients had distant organ metastases at diagnosis (with the most common site being the lung). At diagnosis, only a third of patients were in stage I. The same study calculated a median overall survival of 57.2 months, with 5- and 10-year survival rates of 50 and 38%, respectively. Patients with localized disease had better 5- and 10 years survival rates than those with regional dissemination (82.4%, respectively, 68.8% vs. 44%, respectively, 32.5%) and none of the patients with distant dissemination survived at 5 years [8]. In addition to regional and distant dissemination, there are other prognostic factors associated with the reduction in specific disease-free survival (DFS) and overall survival (OS) for patients with CCS. A diminished DFS was associated with tumors larger than 5 cm (median DFS, 7.5 vs. 25.5 months, p = 0.0043), positive surgical margins (median DFS, 3.5 vs. 13 months, p = 0.0233), and a neutrophile–lymphocyte ratio greater than 2.73 (median DFS, 7.5 vs. 25.5, p = 0.0009). Similar reduced OS rates were associated with a tumor size larger than 5 cm (median OS, 23.5 vs. 63 months, p = 0.0075), positive surgical margins (median OS, 21.5 vs. 63 months, p = 0.0101), a neutrophile–lymphocyte ratio greater than 2.73 (median OS, 26 vs. 85 months, p = 0.0126), a lymphocyte–monocyte ratio smaller than 4.2 (median OS, 26 vs. 85 months, p = 0.0445) and a thrombocyte–lymphocyte greater than 103.89 (median OS, 26 vs. 85 months, p = 0.0147) [8].
Given the rarity of this tumor, there is not a standardized treatment. Radical surgery is essential in the treatment of CCS both for the primary tumor and for recurrence or metastasis. Although complex personalized therapies are available to all citizens based on national health insurance programs [9,10], unfortunately, for this kind of tumor personalized medicine has very little to offer except surgery. Given the rarity of this tumor, there is not a standardized treatment for CCS. Neither ESMO nor NCCN guidelines discuss CCS separately, and only offer general principles for treatment for all soft-tissue sarcomas. Radical surgery is the “gold-standard” in the treatment of CCS both for the primary tumor and for recurrences or metastasis. For a favorable prognosis, early diagnosis followed by surgical treatment with a radical approach is necessary [11]. There are no studies which evaluate the benefits of chemo- and radiotherapy in CCS patients, either in neoadjuvant or adjuvant settings. Adjuvant chemoradiotherapy may improve DFS, but without affecting OS. Systemic therapy is used for unresectable or metastatic cases and is anthracycline-based. Radiation therapy is of little use in gastrointestinal CCS.
As for targeted therapies, none are currently approved for human use and no specific therapeutic agents directly target EWSR1-CREB1 and EWSR1-ATF1 fusion proteins. However, there are a few small studies investigating IGF1R inhibitors for tumors exhibiting EWSR1 fusion proteins [12,13]. EWSR1-ATF1 fusion proteins have been reported to upregulate the expression of MET [14] and MiTF [15], a transcription factor that has been shown to drive MET expression [16,17]. This raises the possibility of using the antibody AMG102 (which suppresses MET signaling) for treating CCS. Using sunitinib for patients with CCS harboring EWSR1-ATF1 fusion has been shown to elicit a therapeutic response, but it is not clear if other genetic alterations are present [18,19]. Combination treatment with crizotinib and pazopanib can determine a durable partial response in a patient with metastatic GNET tumors harboring EWSR1-CREB1 fusion by an unknown mechanism [20]. All these represent potential directions for future research, together with potential therapies targeting STAG2 and MYC gene alterations. As can be observed in our patient NGS’s response, several other genetic alterations were identified but thus far their significance is still unknown; it could possibly represent ways of targeting SCC.

2. Case Presentation

A 44-year-old Caucasian female, with known hereditary predisposition to digestive neoplasms (the mother had colon neoplasm and the father had gallbladder neoplasm), asymptomatic, was diagnosed with mild iron-deficiency anemia on routine checkup in August 2018. In response, endoscopic exploration was ordered and both gastroduodenoscopy and colonoscopy results were found to be normal. Given the oncologic history in the patient’s family, an abdominal tomography scan and entero-MRI (magnetic resonance imaging) were performed, revealing a nonspecific slightly thickened jejunal parietal area without signs of upstream stasis, and no obvious tumors. Digestive tumoral markers were examined and found to be normal. As a result, the patient was scheduled for periodic follow-up.
In the subsequent period, the patient experienced mild diffuse recurrent abdominal pain that did not significantly impact daily routine or sleep quality, and which occurred every 2–3 months.
In May 2020, the patient became abruptly symptomatic, presenting with predominantly nocturnal bilious vomiting, followed by colicky pain. These symptoms recurred every 3–4 days, accompanied by repetitive episodes of belching, hiccups, a sensation of fullness, and diffuse abdominal gurgling, more pronounced in the upper abdominal region. The patient also reported a progressive weight loss of approximately 12 kg in one month in the context of voluntary limitation of food and liquid intake. The symptoms suggested an intermittent ileus and grew in intensity over time. The patient underwent multiple gastroenterology consultations, and the symptoms prompted the performance of an entero-MRI, which revealed a high intestinal obstruction due to a tight stenosis at the level of a jejunal loop (Figure 1). The presence of an invagination was maintained throughout the examination at the level of the described stenotic lesion. Small satellite lymph node images measuring 7/5 mm were also observed. Biochemical investigations revealed hypopotassemia and a slight coagulation deficiency. A complete panel of tumoral markers (consisting of CA19-9, CEA, CA125, CA15-3, CA72-4, AFP, NSE, and SCC) were requested and found to be within normal limits.
In these conditions, with the diagnosis of intestinal obstruction, an emergency surgical intervention was performed. Upon entering the peritoneal cavity, the following were observed: dilated small intestinal loops with a diameter of approximately 5 cm, thickened jejunal walls, and liquid content up to the level of a jejunal stenosis located about 80 cm from the duodeno-jejunal angle and approximately 120 cm from the ileocecal valve. Additionally, numerous lymph nodes with a maximum diameter of 1.5–2 cm were noted along the course of the vascular bundle associated with the affected loop, without definitive macroscopic cancer characteristics (Figure 2).
Under these circumstances, a segmental enterectomy was performed with isolation of the vascular bundle supplying the affected loop at its origin and lymphatic clearance. The resection was carried out within oncological safe limits, with a 10 cm margin on each side of the stenosis of unspecified etiology. The restoration of digestive continuity was achieved through a mechanically assisted side-to-side entero-enteral anastomosis, followed by closure of the mesenteric gap.
The patient was discharged after a favorable postoperative course, with the complete resumption of intestinal transit on the 5th postoperative day, in a satisfactory general condition, afebrile, and with balanced hemodynamic and respiratory status.
The resected specimen (bowel segment and mass) was analyzed. Macroscopically, the pathologist noted a fragment of the small intestine with a dilated proximal portion and an area of vegetative tumor aspect measuring 1.5/1.5/1.2 cm in depth, covered with focally ulcerated, congested mucosa. On section, the tumor had a white color, firm consistency, and predominantly submucosal location with ulceration of the serosa. It covered approximately 30% of the lumen. Additionally, the resected triangular mesenteric portion, measuring 8/8 cm, had numerous small-sized lymph nodes.
Microscopy revealed the resection with clear proximal and distal margins. The intestinal wall showed tumor proliferation with epithelioid and spindle-shaped cells, with alveolar, solid, or storiform architecture. No necrosis was highlighted. Tumor emboli were detected in capillary-caliber vessels of the submucosa. The submitted surgical specimen contained 23 intact lymph nodes (chronic nonspecific lymphadenitis with follicular hyperplasia) and 1 lymph node with a small area of hypocellular fibrosis.
Immunohistochemistry: vimentine positive, CD99, CD15 and CD56 positive, S100 and SOX10 positive, Synaptophisin positive, negative for HMB45, MART-1, Cytokeratine, Desmin, Actin, DOG-1, CD34, CDX-2, CKIT/CD117; Ki67 35%.
Histologic and immunohistochemical findings are summarized in Figure 3.
The definitive diagnosis and further therapeutic approach were determined concomitantly with the histopathological diagnosis. The histopathological results and immunohistochemical profile support the diagnosis of gastrointestinal malignant neuroectodermal tumor/clear cell sarcoma of the gastrointestinal tract (CCS).
The case was then forwarded to an oncologist for further treatment and follow-up. Given that this diagnosis is a rare one, with few cases described in the literature and limited information about therapeutic approaches for adjuvant therapy, a PET-CT was recommended to determine the actual extent of the disease, and revealed no additional tumoral sites. Additionally, a repeat of the immunohistochemistry and analyses of tissue by next generation sequencing (NGS) or Foundation One were recommended and performed. Furthermore, the patient was advised to seek consultation at other international institutes specializing in sarcomas/rare diseases for case analysis and therapeutic guidance.
Genetic testing: microsatellite stable; tumor mutational burden 4 Muts/Mb; BRAF mutations absent; NGS-based assay result showed alteration to the MYC gene, alteration of the EWSR1 gene, with EWSR1-CREB1 fusion present, and alteration to the STAG2 gene. Further, more genetic variants of unknown significance (VUS) were detected in this patient’s tumor. These variants may not have been adequately characterized in the scientific literature at the time this report was issued, and/or the genomic context of these alterations makes their significance unclear, and yet it was decided to include them in this report in the event that they become clinically meaningful in the future. BCOR-V679I; CDK12-P1257del, CREBBP-S128C; ERBB3 rearrangement, IRS2-K1170R; KDM5C-R1435C; MLL2-L4077F; SMO-Q745R.
The patient received secondary medical opinions from multiple international medical facilities specializing in sarcomas/rare diseases in Turkey, the USA, Austria, Greece, and Spain. No clinic provided an indication for adjuvant treatment; the only recommendation was regular follow-up.
The CT scans and MRIs during the first two years of follow-up did not detect any signs of tumor recurrence or metastasis. However, 30 months after primary treatment, the patient developed a unique liver metastasis which was treated by atypical hepatic resection. Similarly, at 46 months, her topography scan showed liver nodules suggestive for secondary implants and some peritoneal nodules in the vicinity of the cecum. The patient underwent laparotomy, and multiple atypical hepatic resections with right hemicolectomy were performed. The pericolic peritoneal nodules were also surgically removed. The postoperative histopathological findings showed aspects compatible with clear cell sarcoma in pericolic nodules (thus confirming peritoneal implants) and in two of the four liver nodules resected. The postoperative course of the patient was marked by a low-flow biliary fistula resulting in a small perihepatic collection which was drained under tomographic guidance. Afterwards, the evolution of the patient was uneventful.
Given the fact that the disease had progressed, systemic therapy was considered upon discussion of the case in a multidisciplinary tumor-board and will be administered to the patient. Regular follow-ups are scheduled.

3. Discussion

In the light of our case, we conducted an extensive search of relevant scientific publications in PubMed and Embase databases and performed a review aimed at highlighting the rarity of this type of tumor and the diagnostic and treatment challenges it raises. All peer-reviewed research and conference papers that describe a CCS with a gastrointestinal localization of tumor and genetic conformation were considered eligible for inclusion. We excluded all reports of non-digestive tumors and reported cases in which FISH or RT-PCR for EWSR1-ATF1 or EWSR1-CREB1 translocations were not performed, thus not allowing for certain diagnostics. We also excluded references that were not available in English, at least in abstract, but included references in all other languages which also had an abstract in English. We searched PubMed from inception until 15 May 2024 using relevant keywords connected by appropriate Boolean operators under the following syntax: (clear cell sarcoma) AND (gastrointestinal OR gastro-intestinal OR abdominal OR intra-abdominal). A similar search strategy was used for Embase search. Additional references were found through a rigorous citation search.
After this search, we obtained 305 results in PubMed and 136 results in Embase. An additional 23 records were identified via reference screening. All records were written between 1976 and 2024. All records retrieved were electronically screened and duplications were removed. Subsequently, two independent reviewers screened the records and excluded records that did not meet our inclusion criteria, had irrelevant focus, or presented cases already included. Discrepancies were solved via group discussion and a senior reviewer’s opinion was taken into consideration in case of disagreement. In total, 41 records were included in this systematic review. Table 1 summarizes the records included.
This study contains a qualitative but not a quantitative summary of the findings, because of the expected high heterogeneity of the articles included. Also, most studies were case reports or very small series of cases. Outcomes were reported in very few patients and, as a result, a proper statistical analysis was impossible.
The male/female ratio was 33/29. The average age was 40.84 years (with extremes of 10–85). Most cases reported small bowel tumors, followed by colonic, gastric, and pancreatic cancers. All cancers were S100 positive, but the majority had negative melanocytic markers (HMB-45, Melan-A). The clear cell sarcomas in the literature had different immunohistochemical signatures, but they all exhibited a translocation or a rearrangement of the Ewing sarcoma breakpoint region 1. In cases where data regarding the outcomes were available, we observed a high aggression of this type of tumor with a tendency to local recurrence and systemic dissemination.

4. Conclusions

Intra-abdominal CCS is an extremely rare tumor, most often misdiagnosed as a MM due to the clinical, histological, and immunohistochemical similarities between the two forms. Our case highlights the difficulty of the diagnosis, which requires firstly that the physician is aware of this pathology and secondly requires very specific genetic testing which is not always available in every institution (in our case, genetic testing was performed outside the country).
Also, the case presented highlights the limited therapeutic options; radical surgery remains the therapeutic “gold-standard” for both primary tumor and recurrent/metastatic disease. Our case demonstrates that a correct surgical technique, with excision with clear margins, can yield good results, as evidenced by a long-term follow-up without tumor recurrence or distant metastasis despite the lack of adjuvant therapy.
In the case of our patient, extensive genetic testing was available which, besides confirming the diagnosis, did in fact show multiple other genetic variants of unknown significance. These variants have not been adequately characterized in the scientific literature at the time of diagnosis, and/or the genomic context of these alterations makes their significance unclear but, in the future, they may become clinically meaningful or provide therapeutic options as possible targetable mutations.

Author Contributions

Conceptualization, E.C. and V.R.; methodology, E.C. and V.R.; investigation, A.G., A.D.P. and S.O.I.; resources, C.C., A.N. and M.A.; writing—original draft preparation, M.N.M. and E.C.; writing—review and editing, E.C., A.G., L.S. and V.R.; supervision, C.C. and V.R.; project administration, L.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Written informed consent has been obtained from the patient to use medical records for educational and scientific purposes, including publication.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We acknowledge the tremendous contribution of Becheanu Gabriel (Pathology Department, Clinical Institute “Fundeni”, Bucharest, Romania) for providing us with a second opinion on histological and immunohistochemical aspects of the case.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

CCSClear cell sarcoma
ATF1Activating transcription factor 1
EWSR1Ewing sarcoma breakpoint region 1
CREB1cAMP responsive element binding protein 1
MMMalignant melanoma
HMB-45Human melanoma black 45 antibody
MiTFMicrophthalmia transcription factor
S100Calcium binding protein
Melan-AMelanocyte antigen
FISHFluorescence in situ hybridization
RT-PCRReal-time polymerase chain reaction
BRAFv-raf murine sarcoma viral oncogene homolog B1
MPNSTMalignant peripheral nerve sheath tumor
GNETGastrointestinal neuroectodermal tumors
CTComputer tomography
MRIMagnetic resonance imaging
CA19-9Carbohydrate antigen 19-9
CEACarcinoembryonic antigen
CA125Carbohydrate antigen 125
CA15-3Carbohydrate antigen 15-3
CA72-4Carbohydrate antigen 72-4
AFPAlpha fetoprotein
NSENeuron-specific enolase
SCCSubfraction of tumor-associated antigens related to squamous cell carcinoma
CD99Cluster of differentiation 99
CD15Cluster of differentiation 15
CD56Cluster of differentiation 56
SOX100Sry-related HMg-Box gene 10
MART-1Melanocyte antigen (also called Melan-A)
DOG-1 gene“Discovered on GIST 1” gene
CD34Cluster of differentiation 34
CDX-2Caudal-type homeobox 2
CKIT/CD117Receptor for tyrosine kinase
Ki67Proliferation index
HEHematoxylin and eosin stain
AbAntibody
PET-CTPositron emission tomography
NGSNext-generation sequencing
VUSGenetic variants of unknown significance
MYC geneMyelocytomatosis oncogene
STAG2 geneStromal antigen 2 gene
DFSDisease-free survival
OSOverall survival
ESMOEuropean Society of Medical Oncology
NCCNNational Comprehensive Cancer Network
METMesenchymal epithelial transition—tyrosine kinase receptor

References

  1. Haouimi, A. Clear cell sarcoma of soft tissue-neck. RadiopaediaOrg 2023. Available online: https://radiopaedia.org/cases/clear-cell-sarcoma-of-soft-tissue-neck (accessed on 15 May 2024). [CrossRef]
  2. Elousrouti, L.T.; Hammas, N.; Elmernissi, F.Z.; Elfatemi, H.; Chbani, L. Clear-Cell Sarcoma with an Unusual Presentation Mimicking Metastatic Melanoma. Cureus 2022, 14, e32010. [Google Scholar] [CrossRef]
  3. Cassalia, F.; Cavallin, F.; Danese, A.; Del Fiore, P.; Di Prata, C.; Rastrelli, M.; Fortina, A.B.; Mocellin, S. Soft Tissue Sarcoma Mimicking Melanoma: A Systematic Review. Cancers 2023, 15, 3584. [Google Scholar] [CrossRef]
  4. Kazakos, C.J.; Galanis, V.G.; Giatromanolaki, A.; Verettas, D.-A.J.; Sivridis, E. Clear cell sarcoma of the scapula. A case report and review of the literature. World J. Surg. Oncol. 2006, 4, 48. [Google Scholar] [CrossRef]
  5. Suehara, Y.; Yazawa, Y.; Hitachi, K.; Terakado, A. Clear cell sarcoma arising from the chest wall: A case report. J. Orthop. Sci. 2004, 9, 171–174. [Google Scholar] [CrossRef]
  6. Clark, M.; Johnson, M.; Thway, K.; Fisher, C.; Thomas, J.; Hayes, A. Clear cell sarcoma (melanoma of soft parts): The Royal Marsden Hospital experience. Eur. J. Surg. Oncol. 2008, 34, 800–804. [Google Scholar] [CrossRef]
  7. Gonzaga, M.I.; Grant, L.; Curtin, C.; Gootee, J.; Silberstein, P.; Voth, E. The epidemiology and survivorship of clear cell sarcoma: A National Cancer Database (NCDB) review. J. Cancer Res. Clin. Oncol. 2018, 144, 1711–1716. [Google Scholar] [CrossRef] [PubMed]
  8. Li, A.-B.; Jiang, B.-J.; Wang, H.-H.; Yang, Y.-S.; Zhang, X.-B.; Lan, G.-H.; Shu, W.-B. Prognostic Factors for Survival in Patients with Clear Cell Sarcoma: An Analysis of the Surveillance, Epidemiology, and End Results (SEER) Database. Med. Sci. Monit. 2019, 25, 6950–6956. [Google Scholar] [CrossRef]
  9. Simion, L.; Rotaru, V.; Cirimbei, C.; Gales, L.; Stefan, D.-C.; Ionescu, S.-O.; Luca, D.; Doran, H.; Chitoran, E. Inequities in Screening and HPV Vaccination Programs and Their Impact on Cervical Cancer Statistics in Romania. Diagnostics 2023, 13, 2776. [Google Scholar] [CrossRef] [PubMed]
  10. Simion, L.; Rotaru, V.; Cirimbei, C.; Stefan, D.-C.; Gherghe, M.; Ionescu, S.; Tanase, B.C.; Luca, D.C.; Gales, L.N.; Chitoran, E. Analysis of Efficacy-to-Safety Ratio of Angiogenesis-Inhibitors Based Therapies in Ovarian Cancer: A Systematic Review and Meta-Analysis. Diagnostics 2023, 13, 1040. [Google Scholar] [CrossRef] [PubMed]
  11. Rodriguez-Martin, M.; Saez-Rodriguez, M.; Esquivel, B.; Gonzalez, R.S.; Cabrera, A.N.; Herrera, A.M. Clear cell sarcoma: A case mimicking primary cutaneous malignant melanoma. Indian J. Dermatol. 2009, 54, 168–172. [Google Scholar] [CrossRef]
  12. Erkizan, H.V.; Kong, Y.; Merchant, M.; Schlottmann, S.; Barber-Rotenberg, J.S.; Yuan, L.; Abaan, O.D.; Chou, T.-H.; Dakshanamurthy, S.; Brown, M.L.; et al. A small molecule blocking oncogenic protein EWS-FLI1 interaction with RNA helicase A inhibits growth of Ewing's sarcoma. Nat. Med. 2009, 15, 750–756. [Google Scholar] [CrossRef]
  13. Li, Y.-J.; Zhao, X.; Vecchiarelli-Federico, L.M.; Datti, A.; Cheng, Y.; Ben-David, Y. Drug-mediated inhibition of Fli-1 for the treatment of leukemia. Blood Cancer J. 2012, 2, e54. [Google Scholar] [CrossRef]
  14. Davis, I.J.; McFadden, A.W.; Zhang, Y.; Coxon, A.; Burgess, T.L.; Wagner, A.J.; Fisher, D.E. Identification of the receptor tyrosine kinase c-Met and its ligand, hepatocyte growth factor, as therapeutic targets in clear cell sarcoma. Cancer Res 2010, 70, 639–645. [Google Scholar] [CrossRef]
  15. Davis, I.J.; Kim, J.J.; Ozsolak, F.; Widlund, H.R.; Rozenblatt-Rosen, O.; Granter, S.R.; Du, J.; Fletcher, J.A.; Denny, C.T.; Lessnick, S.L.; et al. Oncogenic MITF dysregulation in clear cell sarcoma: Defining the MiT family of human cancers. Cancer Cell 2006, 9, 473–484. [Google Scholar] [CrossRef]
  16. McGill, G.G.; Haq, R.; Nishimura, E.K.; Fisher, D.E. c-Met expression is regulated by Mitf in the melanocyte lineage. J. Biol. Chem. 2006, 281, 10365–10373. [Google Scholar] [CrossRef]
  17. Beuret, L.; Flori, E.; Denoyelle, C.; Bille, K.; Busca, R.; Picardo, M.; Bertolotto, C.; Ballotti, R. Up-regulation of MET expression by alpha-melanocyte-stimulating hormone and MITF allows hepatocyte growth factor to protect melanocytes and melanoma cells from apoptosis. J. Biol. Chem. 2007, 282, 14140–14147. [Google Scholar] [CrossRef]
  18. Stacchiotti, S.; Grosso, F.; Negri, T.; Palassini, E.; Morosi, C.; Pilotti, S.; Gronchi, A.; Casali, P.G. Tumor response to sunitinib malate observed in clear-cell sarcoma. Ann. Oncol. 2010, 21, 1130–1131. [Google Scholar] [CrossRef] [PubMed]
  19. Tazzari, M.; Palassini, E.; Vergani, B.; Villa, A.; Rini, F.; Negri, T.; Colombo, C.; Crippa, F.; Morosi, C.; Casali, P.G.; et al. Melan-A/MART-1 immunity in a EWS-ATF1 translocated clear cell sarcoma patient treated with sunitinib: A case report. BMC Cancer 2015, 15, 58. [Google Scholar] [CrossRef] [PubMed]
  20. Subbiah, V.; Holmes, O.; Gowen, K.; Spritz, D.; Amini, B.; Wang, W.-L.; Schrock, A.B.; Meric-Bernstam, F.; Zinner, R.; Piha-Paul, S.; et al. Activity of c-Met/ALK Inhibitor Crizotinib and Multi-Kinase VEGF Inhibitor Pazopanib in Metastatic Gastrointestinal Neuroectodermal Tumor Harboring EWSR1-CREB1 Fusion. Oncology 2016, 91, 348–353. [Google Scholar] [CrossRef] [PubMed]
  21. Donner, L.R.; Trompler, R.A.; Dobin, S. Clear cell sarcoma of the ileum: The crucial role of cytogenetics for the diagnosis. Am. J. Surg. Pathol. 1998, 22, 121–124. [Google Scholar] [CrossRef] [PubMed]
  22. Fukuda, T.; Kakihara, T.; Baba, K.; Yamaki, T.; Yamaguchi, T.; Suzuki, T. Clear cell sarcoma arising in the transverse colon. Pathol. Int. 2000, 50, 412–416. [Google Scholar] [CrossRef]
  23. Pauwels, P.; Debiec-Rychter, M.; Sciot, R.; Vlasveld, T.; Butter, B.D.; Hagemeijer, A.; Hogendoorn, P.C.W. Clear cell sarcoma of the stomach. Histopathology 2002, 41, 526–530. [Google Scholar] [CrossRef] [PubMed]
  24. Zambrano, E.; Reyes-Mugica, M.; Franchi, A.; Rosai, J. An osteoclast-rich tumor of the gastrointestinal tract with features resembling clear cell sarcoma of soft parts: Reports of 6 cases of a GIST simulator. Int. J. Surg. Pathol. 2003, 11, 75–81. [Google Scholar] [CrossRef] [PubMed]
  25. Achten, R.; Debiec-Rychter, M.; De Wever, I.; Sciot, R. An unusual case of clear cell sarcoma arising in the jejunum highlights the diagnostic value of molecular genetic techniques in establishing a correct diagnosis. Histopathology 2005, 46, 472–474. [Google Scholar] [CrossRef] [PubMed]
  26. Venkataraman, G.; Quinn, A.M.; Williams, J.; Hammadeh, R. Clear cell sarcoma of the small bowel: A potential pitfall. APMIS 2005, 113, 716–719. [Google Scholar] [CrossRef] [PubMed]
  27. Covinsky, M.; Gong, S.; Rajaram, V.; Perry, A.; Pfeifer, J. EWS-ATF1 fusion transcripts in gastrointestinal tumors previously diagnosed as malignant melanoma. Hum. Pathol. 2005, 36, 74–81. [Google Scholar] [CrossRef] [PubMed]
  28. Taminelli, L.; Zaman, K.; Gengler, C.; Peloponissios, N.; Bouzourene, H.; Coindre, J.-M.; Hostein, I.; Guillou, L. Primary clear cell sarcoma of the ileum: An uncommon and misleading site. Virchows Arch. 2005, 447, 772–777. [Google Scholar] [CrossRef] [PubMed]
  29. Friedrichs, N.; Testi, M.A.; Moiraghi, L.; Modena, P.; Paggen, E.; Plötner, A.; Wiechmann, V.; Mantovani-Löffler, L.; Merkelbach-Bruse, S.; Buettner, R.; et al. Clear cell sarcoma-like tumor with osteoclast-like giant cells in the small bowel: Further evidence for a new tumor entity. Int. J. Surg. Pathol. 2005, 13, 313–318. [Google Scholar] [CrossRef] [PubMed]
  30. Huang, W.; Zhang, X.; Li, D.; Chen, J.; Meng, K.; Wang, Y.; Lu, Z.; Zhou, X. Osteoclast-rich tumor of the gastrointestinal tract with features resembling those of clear cell sarcoma of soft parts. Virchows Arch. 2005, 448, 200–203. [Google Scholar] [CrossRef]
  31. Antonescu, C.R.; Nafa, K.; Segal, N.H.; Cin, P.D.; Ladanyi, M. EWS-CREB1: A Recurrent Variant Fusion in Clear Cell Sarcoma—Association with Gastrointestinal Location and Absence of Melanocytic Differentiation. Clin. Cancer Res. 2006, 12, 5356–5362. [Google Scholar] [CrossRef]
  32. Granville, L.; Hicks, J.; Popek, E.; Dishop, M.; Tatevian, N.; Lopez-Terrada, D. Visceral Clear Cell Sarcoma of Soft Tissue with Confirmation by EWS-ATF1 Fusion Detection. Ultrastruct. Pathol. 2006, 30, 111–118. [Google Scholar] [CrossRef] [PubMed]
  33. Comin, C.E.; Novelli, L.; Tornaboni, D.; Messerini, L. Clear cell sarcoma of the ileum: Report of a case and review of literature. Virchows Arch. 2007, 451, 839–845. [Google Scholar] [CrossRef]
  34. Abdulkader, I.; Cameselle-Teijeiro, J.; de Alava, E.; Ruiz-Ponte, C.; Used-Aznar, M.M.; Forteza, J. Intestinal clear cell sarcoma with melanocytic differentiation and EWS [corrected] rearrangement: Report of a case. Int. J. Surg. Pathol. 2008, 16, 189–193. [Google Scholar] [CrossRef] [PubMed]
  35. Lyle, P.L.; Amato, C.M.; Fitzpatrick, J.E.; Robinson, W.A. Gastrointestinal melanoma or clear cell sarcoma? Molecular evaluation of 7 cases previously diagnosed as malignant melanoma. Am. J. Surg. Pathol. 2008, 32, 858–866. [Google Scholar] [CrossRef]
  36. Lagmay, J.P.; Ranalli, M.; Arcila, M.; Baker, P. Clear cell sarcoma of the stomach. Pediatr. Blood Cancer 2009, 53, 214–216. [Google Scholar] [CrossRef]
  37. Joo, M.; Chang, S.H.; Kim, H.; Gardner, J.M.; Ro, J.Y. Primary gastrointestinal clear cell sarcoma: Report of 2 cases, one case associated with IgG4-related sclerosing disease, and review of literature. Ann. Diagn. Pathol. 2009, 13, 30–35. [Google Scholar] [CrossRef]
  38. Terazawa, K.; Otsuka, H.; Morita, N.; Yamashita, K.; Nishitani, H. Clear-cell sarcoma of the small intestine detected by FDG-PET/CT during comprehensive examination of an inflammatory reaction. J. Med. Investig. 2009, 56, 70–75. [Google Scholar] [CrossRef]
  39. Shenjere, P.; Salman, W.D.; Singh, M.; Mangham, D.C.; Williams, A.; Eyden, B.P.; Howard, N.; Knight, B.; Banerjee, S.S. Intra-abdominal clear-cell sarcoma: A report of 3 cases, including 1 case with unusual morphological features, and review of the literature. Int. J. Surg. Pathol. 2011, 20, 378–385. [Google Scholar] [CrossRef] [PubMed]
  40. Balkaransingh, P.; Saad, S.A.; Govil, S.C.; Thind, P.K.; Ballance, C.M.; Weiss, A.R. Clear cell sarcoma of the gastrointestinal tract presenting as a second malignant neoplasm following neuroblastoma in infancy. Pediatr. Blood Cancer 2011, 58, 481–482. [Google Scholar] [CrossRef]
  41. Yang, J.C.; Chou, A.J.; Oeffinger, K.C.; La Quaglia, M.P.; Wolden, S.L. Clear cell sarcoma of the gastrointestinal tract after very low-dose therapeutic radiation therapy: A case report. J. Pediatr. Surg. 2012, 47, 1943–1945. [Google Scholar] [CrossRef] [PubMed]
  42. Stockman, D.L.; Miettinen, M.; Suster, S.; Spagnolo, D.M.; Dominguez-Malagon, H.; Hornick, J.L.; Adsay, V.; Chou, P.M.; Amanuel, B.M.; VanTuinen, P.; et al. Malignant gastrointestinal neuroectodermal tumor: Clinicopathologic, immunohistochemical, ultrastructural, and molecular analysis of 16 cases with a reappraisal of clear cell sarcoma-like tumors of the gastrointestinal tract. Am. J. Surg. Pathol. 2012, 36, 857–868. [Google Scholar] [CrossRef]
  43. Suárez-Vilela, D.; Izquierdo, F.M.; Tojo-Ramallo, S.; Riera-Velasco, J.R.; Escobar-Stein, J. Malignant gastrointestinal neuroectodermal tumor showing overlapped immunophenotype with synovial sarcoma: CD99 and SOX10 antibodies are useful in differential diagnosis. Am. J. Surg. Pathol. 2012, 36, 1905–1908. [Google Scholar] [CrossRef] [PubMed]
  44. D’amico, F.E.; Ruffolo, C.; Romeo, S.; Massani, M.; Tos, A.P.D.; Bassi, N. Clear cell sarcoma of the ileum: Report of a case and review of the literature. Int. J. Surg. Pathol. 2011, 20, 401–406. [Google Scholar] [CrossRef]
  45. Lasithiotakis, K.; Protonotarios, A.; Lazarou, V.; Tzardi, M.; Chalkiadakis, G. Clear cell sarcoma of the jejunum: A case report. World J. Surg. Oncol. 2013, 11, 17. [Google Scholar] [CrossRef] [PubMed]
  46. Huang, H.; Liu, Q.; Bu, H.; Chen, M.; Chen, H.; Lin, Y.; Zhang, H. Clear cell sarcoma of gastrointestinal tract: Clinicopathologic analyses and review of literatures. Chin. J. Clin. Exp. Pathol. 2014, 30, 383–388. [Google Scholar] [CrossRef]
  47. Kong, J.; Li, N.; Wu, S.; Guo, X.; Gu, C.; Feng, Z. Malignant gastrointestinal neuroectodermal tumor: A case report and review of the literature. Oncol. Lett. 2014, 8, 2687–2690. [Google Scholar] [CrossRef]
  48. Liu, C.; Ren, Y.; Li, X.; Cao, Y.; Chen, Y.; Cui, X.; Li, L.; Li, F. Absence of 19 known hotspot oncogenic mutations in soft tissue clear cell sarcoma: Two cases report with review of the literature. Int. J. Clin. Exp. Pathol. 2014, 7, 5242–5249. [Google Scholar] [PubMed]
  49. Thway, K.; Judson, I.; Fisher, C. Clear Cell Sarcoma-Like Tumor of the Gastrointestinal Tract, Presenting as a Second Malignancy after Childhood Hepatoblastoma. Case Rep. Med. 2014, 2014, 984369. [Google Scholar] [CrossRef]
  50. Huang, J.; Luo, R.-K.; Du, M.; Zeng, H.-Y.; Chen, L.-L.; Ji, Y. Clear cell sarcoma of the pancreas: A case report and review of literature. Int. J. Clin. Exp. Pathol. 2015, 8, 2171–2175. [Google Scholar]
  51. Yegen, G.; Güllüoğlu, M.; Mete, Ö.; Önder, S.; Kapran, Y. Clear cell sarcoma-like tumor of the gastrointestinal tract: A case report and review of the literature. Int. J. Surg. Pathol. 2014, 23, 61–67. [Google Scholar] [CrossRef] [PubMed]
  52. Raskin, G.A.; Pozharisski, K.M.; Iyevleva, A.G.; Rikov, I.V.; Orlova, R.V.; Imyanitov, E.N. Unusual Clinical Presentation of Gastrointestinal Clear Cell Sarcoma. Gastrointest. Tumors 2015, 2, 83–88. [Google Scholar] [CrossRef] [PubMed]
  53. Moslim, M.A.; Falk, G.A.; Cruise, M.; Morris-Stiff, G. Simultaneous Clear Cell Sarcomas of the Duodenum and Jejunum. Case Rep. Med. 2016, 2016, 1534029. [Google Scholar] [CrossRef] [PubMed]
  54. Ardakani, A.G.; Boyle, D.; Elton, C. Gastrointestinal clear cell sarcoma-like tumour of the ascending colon. Ann. R. Coll Surg. Engl. 2016, 98, e37–e39. [Google Scholar] [CrossRef] [PubMed]
  55. Su, H.; Liu, W.-S.; Ren, W.-H.; Wang, P.; Shi, L.; Zhou, H.-T. Multiple clear-cell sarcomas of small intestine with parotid gland metastasis: A case report. World J. Gastroenterol. 2017, 23, 2258–2265. [Google Scholar] [CrossRef] [PubMed]
  56. Kato, T.; Ichihara, S.; Gotoda, H.; Muraoka, S.; Kubo, T.; Sugita, S.; Hasegawa, T. Imprint cytology of clear cell sarcoma-like tumor of the gastrointestinal tract in the small intestine: A case report. Diagn. Cytopathol. 2017, 45, 1137–1141. [Google Scholar] [CrossRef]
  57. Askan, G.; Kombak, F.E.; Seven, I.E.; Basturk, O. Clear Cell Sarcoma-Like Tumor of the Gastrointestinal Tract. J. Gastrointest. Cancer 2018, 50, 651–656. [Google Scholar] [CrossRef] [PubMed]
  58. Okada, T.; Hirano, Y.; Ishikawa, S.; Kondo, H.; Ishii, T.; Yamaguchi, S. A long-term survivor of clear cell sarcoma-like tumor of the gastrointestinal tract with liver metastasis: A case report. Surg. Case Rep. 2020, 6, 1–9. [Google Scholar] [CrossRef] [PubMed]
  59. Zhu, P.; Zhang, T.; Bi, K.; Wu, Y.; Chen, X.; Zhang, H.; Huang, D.; Zhang, L.; Zeng, Y.; Yi, X. Primary Clear Cell Sarcoma of the Ileum: A Case Report With Next-Generation Sequencing Analysis. Int. J. Surg. Pathol. 2021, 29, 677–684. [Google Scholar] [CrossRef]
  60. Huang, W.-P.; Li, L.-M.; Gao, J.-B. Postoperative multiple metastasis of clear cell sarcoma-like tumor of the gastrointestinal tract in adolescent: A case report. World J. Clin. Cases 2022, 10, 6175–6183. [Google Scholar] [CrossRef]
  61. Njima, M.; Lahbacha, B.; Ben Jabra, S.; Moussa, A.; Bellalah, A.; Ben Abdeljelil, N.; Ben Hammouda, S.; Njim, L.; Hadhri, R.; Zakhama, A. Small Intestine Gastrointestinal Clear Cell Sarcoma: A Case Report and Review of the Literature. J. Investig. Med. High Impact Case Rep. 2024, 12, 23247096231225869. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Entero-MRI which revealed a high intestinal obstruction with multiple distended jejunal loops visible marked by red arrows.
Figure 1. Entero-MRI which revealed a high intestinal obstruction with multiple distended jejunal loops visible marked by red arrows.
Medicina 60 00847 g001
Figure 2. (A) Intraoperative image of the circumferentially thickened jejunal loop, leading to significant dilation upstream of the jejunal loops. (B) Dilated small intestinal loops with a diameter of approximately 5 cm. (C) Intraoperative aspect showing the mesentery of tumoral intestinal loop with enlarged lymph nodes (black arrows) and vascular pedicle identified at the origin from mesenteric artery, and dissected (forceps). (D) Resection specimen—ileal segment with corresponding mesentery.
Figure 2. (A) Intraoperative image of the circumferentially thickened jejunal loop, leading to significant dilation upstream of the jejunal loops. (B) Dilated small intestinal loops with a diameter of approximately 5 cm. (C) Intraoperative aspect showing the mesentery of tumoral intestinal loop with enlarged lymph nodes (black arrows) and vascular pedicle identified at the origin from mesenteric artery, and dissected (forceps). (D) Resection specimen—ileal segment with corresponding mesentery.
Medicina 60 00847 g002
Figure 3. Histologic and immunohistochemical findings. (A) Tumor cells expanding the submucosa (HE, ×25). (B) Tumor cells focally infiltrating the mucosa (HE, ×100). (C) Atypical cells dissecting muscularis propria (HE, ×100). (D) Tumor cells with predominantly epithelioid morphology, prominent nucleoli, and reduced pleomorphism (HE, ×200). (E) Tumor cells with spindle morphology (HE, ×400). (F) Tumor cells are diffusely positive for S100. (G) Tumor cells are diffusely positive for SOX10. (H) Focal positivity for synaptophysin (Anti-Synaptoptophysin Ab, ×400). (I) High proliferation index (Ki67), about 35% (IHC, Anti-Ki67 Ab, ×400).
Figure 3. Histologic and immunohistochemical findings. (A) Tumor cells expanding the submucosa (HE, ×25). (B) Tumor cells focally infiltrating the mucosa (HE, ×100). (C) Atypical cells dissecting muscularis propria (HE, ×100). (D) Tumor cells with predominantly epithelioid morphology, prominent nucleoli, and reduced pleomorphism (HE, ×200). (E) Tumor cells with spindle morphology (HE, ×400). (F) Tumor cells are diffusely positive for S100. (G) Tumor cells are diffusely positive for SOX10. (H) Focal positivity for synaptophysin (Anti-Synaptoptophysin Ab, ×400). (I) High proliferation index (Ki67), about 35% (IHC, Anti-Ki67 Ab, ×400).
Medicina 60 00847 g003
Table 1. Characteristics of all 62 cases already reported in the literature at the time of the study.
Table 1. Characteristics of all 62 cases already reported in the literature at the time of the study.
RecordYearAgeSexLocationS-100HMB-45Melan-AOther IHC FindingsGenetic
Findings
Outcomes
Donner [21]199837MIleum+ND EWSR1-ATF1Liver metastasis at 24 and 36 months
Fukuda [22]200074MColon++ND EWSR1-ATF1Liver metastasis at 9 months
Pauwels [23]200230MStomach++ for vimentin, NSE, CD99
− for cytokeratins, EMA, CD34, CD117, SMA, desmin
EWSR1-ATF1LN and peritoneal metastasis at diagnosis; AWD at 18 months
Zambrano [24]200315FJejunum+− for CD117, CD34EWSR1-ATF1DOD 16 months
Achten [25]200557MJejunum++++ tyrosinase
− for cytokeratins, EMA, chromogranin, CD3, CD117
EWSR1
rearrangements
NS
Venkataraman [26]200521FIleum+− for SMA, tyrosinase, CD34, CD117EWSR1-ATF1NS
Covinsky [27]200547FPancreas+++ EWSR1-ATF1NED after 24 months
85FSmall intestine+++ EWSR1-ATF1DOD at 1 month
Taminelli [28]200535MIleum+++ tyrosinase
− CD117, cytokeratins, EMA, SMA, desmin, CD31, CD34, chromogranin, synaptophysin
EWSR1-ATF1Liver metastasis at 2 months
DOD 15 months
Friedrichs [29]200541MJejunum++ vimentin, beta-catenine, CD68, PDFG-R alfa
− for CD117, CD34, desmin, SMA, chromogranin, synaptophysin, NSE
EWSR1
rearrangements
Liver metastasis at 6 months
Huang [30]200640MStomach+− for CD117, CD34, vimentin, SMA, synaptophysinEWSR1-ATF1NS
Antonescu [31]200681FColon+ EWSR1-CREB1Liver and peritoneal metastasis at 60 months
42FIleum+ EWSR1-CREB1NS
42FIleum+ EWSR1-CREB1Liver and peritoneal metastasis at diagnosis
Granville [32]200516MIleum+ND− for pancytokeratin, CD3, CD34, CD117, EMA, desmin, SMAEWSR1-ATF1DOD 15 months
Comin [33]200731FIleum+− for tyrosinase, cytokeratins, EMASMA, CD34, CD31, CD117, CD99, Synaptophysin, Chromogranin AEWSR1-ATF1NS
Abdulkader [34]200837MJejunum++ND+ PDGF-R alfa, EMA, NSE, vimentine
− CD34, CD117
EWSR1
rearrangement
Liver metastasis at 2 months
Lyle [35]200846MJejunum+++ EWSR1-ATF1NED 7 months
48MCecum+++ EWSR1-ATF1DOD 2 months
60MJejunum+++ EWSR1-ATF1DOD 28 months
62MIleum+++ EWSR1-ATF1DOD 12 months
Lagmay [36]200910FStomach+ EWSR1-ATF1NED 4 months
Joo [37]200960MIleum+ EWSR1
rearrangement
NS
46MJejunum+ EWSR1
rearrangement
NS
Terazawa [38]200920FIleum+NDND EWSR1-ATF1NED at 24 months
Shenjere [39]201153FIleum++ for vimentin, CD57, EMA, MiTF
− for CD34, DOG1, CD99, SMA
EWSR1-ATF1Regional LN metastasis at diagnosis/NED at 7 months
26FSmall and large bowel++ for EMA
− for cytokeratins, CD99, chromogranin, synaptophysin, desmin, CD34
EWSR1-CREB1NS
66MSmall intestine+− for cytokeratins, chromogranin, Synaptophysin, CD56, CD34, CD117, desmin, SMAEWSR1-CREB1Regional LN metastasis at diagnosis/NED
Balkaransingh [40]201115MIleumNDNDND EWSR1
rearrangement
NS
Yang [41]201215MIleum+NDND+ for vimentinEWSR1
rearrangement
Liver metastasis at 12 months
Stockman [42]201230FJejunum++ for SOX10, CD56, NSE, synaptophysinEWSR1-ATF1AWD at 21 months
35MJejunum++ for SOX10, CD56, NSE, sinaptophysinEWSR1-ATF1DOD at 18 months
33MIleum++ for SOX10, CD56
− for synaptophysin, NSE
EWSR1-CREB1AWD at 1.5 months
50FStomach++ for SOX10, synaptophysin
− for CD56, NSE
EWSR1-ATF1AWD at 24 months
20FSmall intestine++ for SOX10, CD56, NSE
− for synaptophysin
EWSR1
rearrangement
NED at 20 months
46MStomach++ for SOX10, CD56
− for synaptophysin, NSE
EWSR1
rearrangement
NS
34FStomach++ for SOX10, CD56
− for synaptophysin, NSE
EWSR1-ATF1DOD at 19 months
77FColon++ for SOX10, CD56, NSE, synaptophysinEWSR1-ATF1DOD at 106 months
17MSmall intestine++ for SOX10, CD56
− for synaptophysin, NSE
EWSR1
rearrangement
NS
60MIleum++ for SOX10, CD56, synaptophysin
− for NSE
EWSR1-CREB1AWD at 36 months
60FJejunum++ for SOX10, CD56
− for synaptophysin
EWSR1-CREB1NED at 41 months
56MStomach++ for SOX10, CD56
− for synaptophysin
EWSR1-CREB1NS
28FSmall intestine++ for SOX10, CD56, NSE, synaptophysinEWSR1
rearrangement
DOD at 23 months
Suárez-Vilela [43]201236FJejunum++ for CD56, vimentin, cytokeratins, EMA
− for CD117, CD99, desmin, SMA, chromogranin, synaptophysin
EWSR1-ATF1NS
D’Amico [44]201269FIleum+ND+ for CD56
− for DOG1, EMA, SMA, CD117, desmin, myogenin
EWSR1
rearrangement
Liver metastasis at 6 months
Lasithiotakis [45]201349FJejunum++ for EMA, synaptophysin
EWSR1-ATF1NED 20 months
Huang [46]201445FColon+− for CD117EWSR1
rearrangement
NS
Kong [47]201417MStomach++ for vimentin
− for CD34, CD117, CD99
EWSR1
rearrangement
NED 10 months
Liu [48] 76MJejunum+ND+ for CD56
− for synaptophysin
EWSR1-ATF1NS
Thway [49]201436MIleum++ for EMA, CD56, NSE
− for SMA, desmin, CD117, DOG1, chromogranin, synaptophysin, CD34
EWSR1-CREB1DOD 7 months; Local recurrence + metastasis of liver, peritoneum, and regional LN at DOD
Huang [50]201536MPancreas++++ for vimentin,MiTF
− for cytokeratins, EMA, desmin, SMA, CD34, CD117, CD99, synaptophysin, chromogranin, CD56, NSE
EWSR1
rearrangement
Liver metastasis at 10 months.
DOD at 10 months
Yegen [51]201525FIleum++ for vimentin, beta-catenin, CD56
− for CD34, CD117, SMA, desmin, chromogranin, synaptophysin
EWSR1
rearrangement
Liver metastasis at diagnosis and at 15 months; Ovarian and peritoneal metastasis at 47 months
Raskin [52]201521MSmall intestine+− for MiTF, synaptophysin, CD56EWSR1-ATF1LN metastasis at diagnosis
Moslim [53]201657MDuodenum and Jejunum (2 tumors)++− for negative for cytokeratins, chromogranin, synaptophysin, desmin, SMA, CD34EWSR1
rearrangement
NED 30 months and then DOD 4 months later due to rapid metastatic progression
Ardakani [54]201622MColon+NS− for SMA, desmin, CD34, CD117, DOG1EWSR1
rearrangement
NS
Su [55]201751MIleum and Jejunum (3 tumors)++++ for vimentin, CD56
− for Synaptophysin, cytokeratins, CD34, CD117, DOG1
EWSR1
rearrangement
NS
Kato [56]201747FColon++ for vimentin, SOX10
− for SMA, CD117, cytokeratin
EWSR1-CREB1NS
Aksan [57]201928MSmall intestine+NS+for SOX10
− for CD117, DOG1, desmin
EWSR1-ATF1Liver and LN metastasis at diagnosis
Okada [58]202038FSmall intestine++ for CD56, synaptophysin
− for desmin, chromogranin, CD34, CD117, SMA
EWSR1
rearrangement
LN metastasis at diagnosis
Liver metastasis at 36 months (surgery); NED at 72 months
Zhu [59]202165MIleum+++ for SOX10, MiTF
− for cytokeratins, EMA, CD117, DOG1, CD34, SMA, desmin, synaptophysin, chromogranin
EWSR1-ATF1NED at 7 months
Huang [60]202216MIleum++ for CD34
− for cytokeratins, CD117, DOG1, desmin, NSE
EWSR1-ATF1DOD at 56 months
Liver, lung, bone, LN, pleural and adrenal metastasis at DOD
Njima [61]202420FIleum++ for SOX10, synaptophysin
− for CD117, DOG1, cytokeratins, CD34, SMA, desmin, chromogranin
ND *NS
Abbreviations: M—male; F—female; S-100—calcium-binding protein; HMB-45—Human Melanoma Black 45 antibody; Melan-A—Melanocyte Antigen; ND—not done; NS—not specified; ND *—not done yet; DOD—date of death; NED—no evidence of disease; AWD—alive with disease; LN—lymph nodes; IHC—immunohistochemistry; NSE—neuron specific enolase; EMA—epithelial membrane antigen; SMA—smooth-muscle actin; CD—cluster of differentiation; PDGF-R—platelet-derived growth factor receptor; SOX10—Sry-related HMg-Box gene 10; DOG1—“Discovered on GIST 1” gene; MiTF—microphthalmia transcription factor; EWSR1—Ewing sarcoma breakpoint region 1; EWSR1-ATF1—translocation between Ewing sarcoma breakpoint region 1 and activating transcription factor 1; EWSR1-CREB1—translocation between Ewing sarcoma breakpoint region 1 and cAMP responsive element binding protein 1.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rotaru, V.; Chitoran, E.; Mitroiu, M.N.; Ionescu, S.O.; Neicu, A.; Cirimbei, C.; Alecu, M.; Gelal, A.; Prie, A.D.; Simion, L. Intestinal Clear Cell Sarcoma—A Case Presentation of an Extremely Rare Tumor and Literature Review. Medicina 2024, 60, 847. https://doi.org/10.3390/medicina60060847

AMA Style

Rotaru V, Chitoran E, Mitroiu MN, Ionescu SO, Neicu A, Cirimbei C, Alecu M, Gelal A, Prie AD, Simion L. Intestinal Clear Cell Sarcoma—A Case Presentation of an Extremely Rare Tumor and Literature Review. Medicina. 2024; 60(6):847. https://doi.org/10.3390/medicina60060847

Chicago/Turabian Style

Rotaru, Vlad, Elena Chitoran, Madalina Nicoleta Mitroiu, Sinziana Octavia Ionescu, Ariana Neicu, Ciprian Cirimbei, Mihnea Alecu, Aisa Gelal, Andra Delia Prie, and Laurentiu Simion. 2024. "Intestinal Clear Cell Sarcoma—A Case Presentation of an Extremely Rare Tumor and Literature Review" Medicina 60, no. 6: 847. https://doi.org/10.3390/medicina60060847

Article Metrics

Back to TopTop