Editorial Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Aug 16, 2024; 12(23): 5299-5303
Published online Aug 16, 2024. doi: 10.12998/wjcc.v12.i23.5299
Essential role of postoperative follow-up in the management of clear cell sarcoma
Zi-Han Zhang, Jin-Tao Guo, Ying Xie, Si-Yu Sun, Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
Jin-Tao Guo, Si-Yu Sun, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shenyang 110004, Liaoning Province, China
ORCID number: Zi-Han Zhang (0000-0001-7636-9400); Jin-Tao Guo (0000-0001-5722-6359); Ying Xie (0000-0001-8226-4242); Si-Yu Sun (0000-0002-7308-0473).
Author contributions: Zhang ZH, Guo JT, Xie Y, Sun SY contributed to this paper; Zhang ZH designed the outline of the manuscript and drafted the article; Guo JT was responsible for the revision of the manuscript; Xie Y contributed to the editing the manuscript; Sun SY was the study supervisor and was responsible for the revision of the manuscript.
Supported by Liaoning Province Applied Basic Research Program Joint Program Project, No. 2022JH2/101500076; Shenyang Young and Middle-aged Science and Technology Innovation Talent Support Program, No. RC200438; Tree Planting Program of Shengjing Hospital, No. M1595; and the Doctoral Start-up Foundation of Liaoning Province, No. 2022-BS-127.
Conflict-of-interest statement: All the authors report having no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Si-Yu Sun, MD, PhD, Chief Doctor, Director, Professor, Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Shenyang 110004, Liaoning Province, China. sun-siyu@163.com
Received: April 7, 2024
Revised: May 18, 2024
Accepted: June 4, 2024
Published online: August 16, 2024
Processing time: 89 Days and 2.8 Hours

Abstract

Clear cell sarcoma (CCS) is a rare melanocytic soft tissue sarcoma known for its propensity to metastasize to the lymph nodes and typically has an unfavorable prognosis. Currently, surgical resection is the primary treatment for localized CCS, while radiotherapy and chemotherapy are preferred for metastatic cases. The roles of adjuvant chemotherapy, radiotherapy, and lymph node dissection are controversial. Although immunotherapy has emerged as a promising avenue in CCS treatment research, there are no established clinical standards for postoperative follow-up. This editorial discusses a recent article by Liu et al, with a focus on current diagnostic modalities, treatment approaches, and the challenging prognosis associated with CCS. Our aim is to underscore the importance of long-term patient follow-up in CCS management.

Key Words: Clear cell carcinoma, Diagnosis, Treatment, Prognosis, Follow-up

Core Tip: Clear cell sarcoma (CCS) is a rare subtype of melanocytic soft tissue sarcoma distinguished by its propensity for metastasis and recurrence. Survival rates markedly diminish in patients with advanced CCS compared with those with early-stage disease. The insidious nature of early occult onset and the limited responsiveness of CCS to chemoradiotherapy underscore the critical importance of routine postoperative follow-up for early detection of recurrent cases, which is crucial for enhancing patient prognosis.



INTRODUCTION

Clear cell sarcoma (CCS) is an uncommon and highly aggressive malignant soft tissue sarcoma (STS) it is characterized by melanocytic differentiation and is believed to originate in tendons and aponeuroses. Due to its insidious onset and slow growth in its early stages, CCS is frequently misdiagnosed as a benign lesion or is identified at an advanced stage. It poses a significant diagnostic challenge. Predominantly affecting the lower extremities, particularly the feet and ankles, CCS exhibits aggressive behavior, marked by local recurrence, lymph node metastasis, and distant metastasis[1,2]. At diagnosis, the majority of patients already present with lymph node or distant metastasis[3], contributing to the generally poor prognosis, with notably lower 5-year survival rates in advanced stages compared to those in early stages. Metastasis primarily occurs in the lungs, followed by the bones and brain[3]. While CCS of the gastrointestinal system is exceedingly rare[4], clear cell sarcoma-like gastrointestinal tumor (CCSLGT), initially reported by Zambrano et al[5] in 2003, is characterized by robust diffuse immunoreactivity of S-100 protein, negative melanocyte-specific markers, and the presence of osteoclast-like multinucleated giant cells. It primarily affects the walls of the small intestine, stomach, or large intestine. Although only two cases of primary pancreatic CCS have been reported to date[4,6], CCSLGT has a more aggressive phenotype compared with traditional CCS, often presenting with metastasis at diagnosis, particularly to the lymph nodes or liver[7].

Despite advancements in understanding the pathogenesis of STS, agreement on the management of CCS is lacking because it is a rare disease. This editorial sheds light on the latest developments in CCS diagnosis and treatment, underscoring the necessity of postoperative follow-up of patients with CCS.

CURRENT DIAGNOSTIC METHODS FOR CCS

The diagnosis of CCS typically relies on a combination of medical history, clinical presentation, and imaging findings. Many patients are already at an advanced stage at symptom onset. CCSLGT predominantly affects the intestinal wall, manifesting as abdominal pain, intestinal obstruction, and occasionally nonspecific symptoms such as anorexia, night sweats, weight loss, or anemia[7]. Patients with pancreatic CCS commonly present with symptoms such as anorexia, jaundice, or other gastrointestinal symptoms[4,6].

Computed tomography (CT) is a primary diagnostic tool for identifying suspected cases, offering insights into sarcoma size and location[8]. Magnetic resonance imaging (MRI) aids in assessing tumor depth and its relationship with surrounding structures and is needed for preoperative planning. Liu et al[9] reported a rare case of CCS with pancreatic metastasis, elucidating the imaging features of pancreatic CCS. CT imaging revealed a round, mildly enhanced, heterogeneous lesion in the pancreatic tail and MRI depicted a vague boundary between the lesion and pancreatic body, with an equal to slightly lower signal on T1 and slightly higher signal on T2, along with progressive uneven enhancement. Given CCS's propensity for hematological or lymphatic metastasis, staging typically involves preoperative assessment with whole-body CT, positron emission tomography-CT, or bone scintigraphy[10]. Recent reports underscore the possibility of peripheral blood transmission of CCS, highlighting the utility of blood smears for rapid diagnosis[11].

A histological examination is imperative to confirm the diagnosis of CCS[12]. Additionally, preoperative confirmation through percutaneous biopsy is essential to ensure negative margins[13]. Multinucleated giant cells that have a wreath-like nuclear appearance are often present in CCS[14]. In contrast, CCSLGT typically lacks conspicuous nucleoli and multinucleated giant cells, replaced instead by CD68-positive multinucleated osteoclast-like giant cells[15]. Immunohistochemical analysis of CCS demonstrates striking similarities to malignant melanoma, commonly expressing S100 protein and melanocyte-specific markers such as HMB-45, melanin-A, and microphthalmia-associated transcription factor (MITF)[16-18]. While S100 protein is expressed in nearly all cases of CCSLGT, melanoma markers (melan A, HMB45) are typically negative[15].

Differential diagnosis is critical in distinguishing CCS from S100 protein-positive and/or melanocyte marker-positive tumors, including malignant melanoma, epithelioid malignant peripheral nerve sheath tumors, melanotic schwannomas, and perivascular epithelioid cell tumors[19]. Despite having. morphological, ultrastructural, and histopathological similarities, CCS and melanoma differ in their molecular tumorigenesis[20]. Recent advancements have used fluorescence in situ hybridization and reverse transcription polymerase chain reaction to confirm CCS and distinguish it from malignant melanoma based on distinct genetic profiles[21]. Approximately 70%–90% of CCS cases are detected by the t(12;22)(q13;q12) translocation, which results in the Ewing sarcoma breakpoint region 1 (EWSR1)/activating transcription factor 1 (ATF1) chimeric gene. This translocation induces MITF expression, facilitating the acquisition of melanocytic characteristics and the expression of melanocyte markers[22]. Another specific mutation, the t(2;22)(q34;q12) translocation leading to EWSR1 and cAMP response element-binding protein (CREB1) fusion, is found in a minority of CCS cases. More recently, EWSR/cAMP responsive element modulator (CREM) fusion was identified in several CCS cases[21]. Ozenberger et al[23] used V5 labeled EWSR1/ATF1 to induce CCS in mice to observe genome distribution and found that CREM may represent an important target gene and cofactor for EWSR1/ATF1 fusion. Notably, BRAF mutations, common in melanoma, are rare in CCS[24].

CURRENT MANAGEMENT OF CCS

Currently, the preferred treatment for localized CCS is complete surgical resection with negative margins, and is often followed by local excision or partial amputation. While standards for metastasis detection remain elusive, radiation or chemotherapy is the preferred treatment modality[3]. The most frequent systemic sarcoma chemotherapy regimens include doxorubicin, sunitinib, gemcitabine, and pazopanib[25]. However, clinical trials evaluating MET inhibitors, such as tivantinib and crizotinib, have demonstrated limited antitumor activity, necessitating further research and improvement of treatment strategies[26,27]. Notably, trabectedin has recently been shown to induce cell cycle arrest in CCS cell lines, inhibit CCS tumor growth in mice, and elicit clinical and radiological tumor responses in patients[28,29].

CCS is generally regarded as unresponsive to radiotherapy and chemotherapy, with conflicting evidence of the survival advantage of adjuvant radiotherapy and chemotherapy[13,19,30,31]. A randomized prospective study by Beane et al[32] indicated that postoperative radiotherapy significantly reduced local recurrence rates in patients with STS, albeit without enhancing overall survival rates. The Japanese Orthopedic Association guidelines advocate for perioperative adjuvant radiotherapy in patients with STS, emphasizing the necessity for meticulous individual case assessment to optimize treatment outcome[33].

Recently, significant attention has been directed towards the development and application of immunotherapies for advanced sarcomas. The fundamental principle involves inhibiting the checkpoints that tumors use as a defense mechanism to evade detection by immune system. The use of immune checkpoints such as cytotoxic T-lymphocyte antigen-4 and programmed cell death-1 (PD-1) in melanoma treatment, and their parallels with CCS, have captured the interest of immunotherapists in the treatment of CCS. Marcrom et al[34] reported a case of recurrent, unresectable CCS with a complete clinical response to pembrolizumab and radiotherapy. This suggests that there is a synergistic effect between PD-1 therapy and radiotherapy. A phase I/II clinical trial conducted by Gordon et al[35] found the combined use of immune checkpoint inhibitors ipilimumab and nivolumab and the tumoricide trabectedin (the SAINT regimen) in early-stage disease significantly enhanced the survival outcomes of patients with advanced sarcoma, with CCS having a favorable responsive. However, large-scale studies are urgently needed to explore the therapeutic potential of immunotherapy in patients with CCS.

The prognosis of patients with CCS remains dismal, with 5- and 10-year survival rates ranging from 47% to 67% and 25% to 41%, respectively[19]. Notably, the 5-year overall survival rates of patients with stage III and IV CCS are substantially lower than those of patients with stage I and II CCS. Approximately one-third of patients with CCS have lymph node metastasis at diagnosis, a prevalence significantly higher than that observed in other soft tissue sarcomas[3]. In a retrospective analysis of 60 cases categorized as CCSLGT, Washimi et al[15] found that the frequency of lymph node metastasis at diagnosis was 62%, which was significantly higher. than the metastasis rate observed in CCS. Previous research has highlighted tumor size, location, and metastasis as key prognostic factors[36]. A 2023 national database study by Fujiwara et al[33] identified primary tumor resection and metastatic lymph node dissection as independent factors associated with improved prognosis.

Moreover, CCS has a recurrence rate as high as 40%[37], with reports of late recurrences occurring after more than 10 years[13]. Fujiwara et al[33] reported cumulative local recurrence rates of 11% at 3 years and 19% at 5 years postoperatively. Tumor size was found to be significantly correlated with local recurrence, in contrast to other soft tissue sarcomas, in which surgical margins and the use of chemotherapy or radiotherapy typically has a more significant role. Nonetheless, the limited patient cohort in this study raises concerns about the reliability of this conclusion, necessitating further investigation.

The high recurrence rate and aggressive nature of CCS underscore the critical importance of comprehensive long-term follow-up for all patients. However, there is currently a lack of published data outlining the optimal routine follow-up strategy for surgically treated patients with localized disease. In the case reported by Liu et al[9], the patient declined regular disease monitoring and examination after the first operation, seeking follow-up only when symptoms manifested, resulting in delayed treatment. This highlights the crucial role of regular postoperative follow-ups in patients with CCS. The clinical practice guidelines established by ESMO-EURACAN-GENTURIS recommend risk assessment and a tailored follow-up strategy based on histological type and tumor grade, size, and location. Additionally, the use of appropriate imaging modalities, such as MRI to detect local recurrence in extremities and superficial recurrence in the trunk and CT to identify lung metastases, are useful for early detection of recurrence[38].

CONCLUSION

Given the rarity of CCS, our understanding of the disease remains partial, necessitating further research to advance our understanding of its diagnosis and management. However, increased rates of metastasis and recurrence underscore the importance of prioritizing postoperative care and devising a structured follow-up regimen for patients. Early identification of at-risk individuals and prompt recognition of recurrent cases stand as crucial measures to improve patient prognosis and increase survival rates.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology & hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade B, Grade C

Novelty: Grade B, Grade C

Creativity or Innovation: Grade B, Grade C

Scientific Significance: Grade B, Grade C

P-Reviewer: Dey T, India; Sahin TT, Türkiye S-Editor: Gong ZM L-Editor: Filipodia P-Editor: Chen YX

References
1.  Bianchi G, Charoenlap C, Cocchi S, Rani N, Campagnoni S, Righi A, Frisoni T, Donati DM. Clear cell sarcoma of soft tissue: a retrospective review and analysis of 31 cases treated at Istituto Ortopedico Rizzoli. Eur J Surg Oncol. 2014;40:505-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
2.  Enzinger FM. Clear-cell sarcoma of tendons and aponeuroses. An analysis of 21 cases. Cancer. 1965;18:1163-1174.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Gonzaga MI, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 32]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
4.  Huang J, Luo RK, Du M, Zeng HY, Chen LL, Ji Y. Clear cell sarcoma of the pancreas: a case report and review of literature. Int J Clin Exp Pathol. 2015;8:2171-2175.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 119]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
6.  Xiang H, Xiang W, Wang L. Primary clear cell-sarcoma of the pancreas: A case report. Asian J Surg. 2023;46:4842-4843.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
7.  Wang J, Thway K. Clear cell sarcoma-like tumor of the gastrointestinal tract: an evolving entity. Arch Pathol Lab Med. 2015;139:407-412.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 53]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
8.  Sabaté JM, Fernández A, Torrubia S, Villanueva A, Monill JM. Clear cell sarcoma of the abdominal wall with peritoneal sarcomatosis: CT features. Eur Radiol. 1999;9:1550-1552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
9.  Liu YJ, Zou C, Wu YY. Metastatic clear cell sarcoma of the pancreas: A rare case report. World J Clin Cases. 2024;12:1448-1453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
10.  Ibrahim RM, Steenstrup Jensen S, Juel J. Clear cell sarcoma-A review. J Orthop. 2018;15:963-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 19]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
11.  Zhang Y, Guo N, Shen N, Li K, Zheng J, Li T, Long F. Circulating tumor cells in clear cell sarcoma. Br J Haematol. 2023;201:1018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
12.  Cortellini F, Carrara S, Fusaroli P. EUS-guided fine-needle biopsy for gastric submucosal tumors: Does one size fit all? Endosc Ultrasound. 2022;11:151-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
13.  Clark MA, Johnson MB, Thway K, Fisher C, Thomas JM, Hayes AJ. Clear cell sarcoma (melanoma of soft parts): The Royal Marsden Hospital experience. Eur J Surg Oncol. 2008;34:800-804.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
14.  Cornillie J, van Cann T, Wozniak A, Hompes D, Schöffski P. Biology and management of clear cell sarcoma: state of the art and future perspectives. Expert Rev Anticancer Ther. 2016;16:839-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 23]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
15.  Washimi K, Takagi M, Hisaoka M, Kawachi K, Takeyama M, Hiruma T, Narimatsu H, Yokose T. Clear cell sarcoma-like tumor of the gastrointestinal tract: A clinicopathological review. Pathol Int. 2017;67:534-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
16.  De Beuckeleer LH, De Schepper AM, Vandevenne JE, Bloem JL, Davies AM, Oudkerk M, Hauben E, Van Marck E, Somville J, Vanel D, Steinbach LS, Guinebretière JM, Hogendoorn PC, Mooi WJ, Verstraete K, Zaloudek C, Jones H. MR imaging of clear cell sarcoma (malignant melanoma of the soft parts): a multicenter correlative MRI-pathology study of 21 cases and literature review. Skeletal Radiol. 2000;29:187-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 57]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
17.  Hocar O, Le Cesne A, Berissi S, Terrier P, Bonvalot S, Vanel D, Auperin A, Le Pechoux C, Bui B, Coindre JM, Robert C. Clear cell sarcoma (malignant melanoma) of soft parts: a clinicopathologic study of 52 cases. Dermatol Res Pract. 2012;2012:984096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
18.  Coindre JM, Hostein I, Terrier P, Bouvier-Labit C, Collin F, Michels JJ, Trassard M, Marques B, Ranchere D, Guillou L. Diagnosis of clear cell sarcoma by real-time reverse transcriptase-polymerase chain reaction analysis of paraffin embedded tissues: clinicopathologic and molecular analysis of 44 patients from the French sarcoma group. Cancer. 2006;107:1055-1064.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 75]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
19.  Ikuta K, Nishida Y, Imagama S, Tanaka K, Ozaki T. The current management of clear cell sarcoma. Jpn J Clin Oncol. 2023;53:899-904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
20.  Potter AJ, Dimitriou F, Karim RZ, Mahar A, Chan C, Long GV, Scolyer RA. Cutaneous clear cell sarcoma with an epidermal component mimicking melanoma. Pathology. 2022;54:369-371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
21.  Segawa K, Sugita S, Aoyama T, Kubo T, Asanuma H, Sugawara T, Ito Y, Tsujiwaki M, Fujita H, Emori M, Hasegawa T. Detection of specific gene rearrangements by fluorescence in situ hybridization in 16 cases of clear cell sarcoma of soft tissue and 6 cases of clear cell sarcoma-like gastrointestinal tumor. Diagn Pathol. 2018;13:73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
22.  Li KK, Goodall J, Goding CR, Liao SK, Wang CH, Lin YC, Hiraga H, Nojima T, Nagashima K, Schaefer KL, Lee KA. The melanocyte inducing factor MITF is stably expressed in cell lines from human clear cell sarcoma. Br J Cancer. 2003;89:1072-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
23.  Ozenberger BB, Li L, Wilson ER, Lazar AJ, Barrott JJ, Jones KB. EWSR1::ATF1 Orchestrates the Clear Cell Sarcoma Transcriptome in Human Tumors and a Mouse Genetic Model. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
24.  Park BM, Jin SA, Choi YD, Shin SH, Jung ST, Lee JB, Lee SC, Yun SJ. Two cases of clear cell sarcoma with different clinical and genetic features: cutaneous type with BRAF mutation and subcutaneous type with KIT mutation. Br J Dermatol. 2013;169:1346-1352.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
25.  Smrke A, Frezza AM, Giani C, Somaiah N, Brahmi M, Czarnecka AM, Rutkowski P, Van der Graaf W, Baldi GG, Connolly E, Duffaud F, Huang PH, Gelderblom H, Bhadri V, Grimison P, Mahar A, Stacchiotti S, Jones RL. Systemic treatment of advanced clear cell sarcoma: results from a retrospective international series from the World Sarcoma Network. ESMO Open. 2022;7:100522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
26.  Schöffski P, Wozniak A, Stacchiotti S, Rutkowski P, Blay JY, Lindner LH, Strauss SJ, Anthoney A, Duffaud F, Richter S, Grünwald V, Leahy MG, Reichardt P, Sufliarsky J, van der Graaf WT, Sciot R, Debiec-Rychter M, van Cann T, Marréaud S, Lia M, Raveloarivahy T, Collette L, Bauer S. Activity and safety of crizotinib in patients with advanced clear-cell sarcoma with MET alterations: European Organization for Research and Treatment of Cancer phase II trial 90101 'CREATE'. Ann Oncol. 2017;28:3000-3008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 56]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
27.  Wagner AJ, Goldberg JM, Dubois SG, Choy E, Rosen L, Pappo A, Geller J, Judson I, Hogg D, Senzer N, Davis IJ, Chai F, Waghorne C, Schwartz B, Demetri GD. Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: results of a multicenter phase 2 trial. Cancer. 2012;118:5894-5902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 122]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
28.  Galera M, Álvarez R, Arregui M, Paniagua M, Álvarez A, González Crisostomo RA, Díazgranados A, Gutiérrez N, Calles A, Agra C. A Clear Cell Sarcoma Case: A Diagnostic and Treatment Challenge, with a Promising Response to Trabectedin. Case Rep Oncol. 2023;16:1542-1550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
29.  Nakai T, Imura Y, Tamiya H, Yamada S, Nakai S, Yasuda N, Kaneko K, Outani H, Takenaka S, Hamada K, Myoui A, Araki N, Ueda T, Itoh K, Yoshikawa H, Naka N. Trabectedin is a promising antitumor agent potentially inducing melanocytic differentiation for clear cell sarcoma. Cancer Med. 2017;6:2121-2130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
30.  Kawai A, Hosono A, Nakayama R, Matsumine A, Matsumoto S, Ueda T, Tsuchiya H, Beppu Y, Morioka H, Yabe H; Japanese Musculoskeletal Oncology Group. Clear cell sarcoma of tendons and aponeuroses: a study of 75 patients. Cancer. 2007;109:109-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 100]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
31.  Mavrogenis A, Bianchi G, Stavropoulos N, Papagelopoulos P, Ruggieri P. Clinicopathological features, diagnosis and treatment of clear cell sarcoma/melanoma of soft parts. Hippokratia. 2013;17:298-302.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Beane JD, Yang JC, White D, Steinberg SM, Rosenberg SA, Rudloff U. Efficacy of adjuvant radiation therapy in the treatment of soft tissue sarcoma of the extremity: 20-year follow-up of a randomized prospective trial. Ann Surg Oncol. 2014;21:2484-2489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 163]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
33.  Fujiwara T, Kunisada T, Nakata E, Mitsuhashi T, Ozaki T, Kawai A. Factors associated with survival in patients with clear cell sarcoma. Bone Joint J. 2023;105-B:1216-1225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
34.  Marcrom S, De Los Santos JF, Conry RM. Complete response of mediastinal clear cell sarcoma to pembrolizumab with radiotherapy. Clin Sarcoma Res. 2017;7:14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 12]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
35.  Gordon EM, Chawla SP, Tellez WA, Younesi E, Thomas S, Chua-Alcala VS, Chomoyan H, Valencia C, Brigham DA, Moradkhani A, Quon D, Srikureja A, Wong SG, Tseng W, Federman N. SAINT: A Phase I/Expanded Phase II Study Using Safe Amounts of Ipilimumab, Nivolumab and Trabectedin as First-Line Treatment of Advanced Soft Tissue Sarcoma. Cancers (Basel). 2023;15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 6]  [Reference Citation Analysis (0)]
36.  Wetterwald L, Riggi N, Kyriazoglou A, Dei Tos G, Dei Tos A, Digklia A. Clear cell sarcoma: state-of-the art and perspectives. Expert Rev Anticancer Ther. 2023;23:235-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
37.  Kunisada T, Nakata E, Fujiwara T, Hosono A, Takihira S, Kondo H, Ozaki T. Soft-tissue sarcoma in adolescents and young adults. Int J Clin Oncol. 2023;28:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
38.  Gronchi A, Miah AB, Dei Tos AP, Abecassis N, Bajpai J, Bauer S, Biagini R, Bielack S, Blay JY, Bolle S, Bonvalot S, Boukovinas I, Bovee JVMG, Boye K, Brennan B, Brodowicz T, Buonadonna A, De Álava E, Del Muro XG, Dufresne A, Eriksson M, Fagioli F, Fedenko A, Ferraresi V, Ferrari A, Frezza AM, Gasperoni S, Gelderblom H, Gouin F, Grignani G, Haas R, Hassan AB, Hecker-Nolting S, Hindi N, Hohenberger P, Joensuu H, Jones RL, Jungels C, Jutte P, Kager L, Kasper B, Kawai A, Kopeckova K, Krákorová DA, Le Cesne A, Le Grange F, Legius E, Leithner A, Lopez-Pousa A, Martin-Broto J, Merimsky O, Messiou C, Mir O, Montemurro M, Morland B, Morosi C, Palmerini E, Pantaleo MA, Piana R, Piperno-Neumann S, Reichardt P, Rutkowski P, Safwat AA, Sangalli C, Sbaraglia M, Scheipl S, Schöffski P, Sleijfer S, Strauss D, Strauss S, Sundby Hall K, Trama A, Unk M, van de Sande MAJ, van der Graaf WTA, van Houdt WJ, Frebourg T, Casali PG, Stacchiotti S; ESMO Guidelines Committee, EURACAN and GENTURIS. Soft tissue and visceral sarcomas: ESMO-EURACAN-GENTURIS Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2021;32:1348-1365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 367]  [Article Influence: 122.3]  [Reference Citation Analysis (0)]