1
|
Argani P, Wangsiricharoen S, Tretiakova M, Liu YJ, Falzarano SM, Collins K, Brimo F, Gross JM, Baraban E, Matoso A, Wakeman K, Corless C, Neff T, Smith BF, Abdel Satir A, Agaimy A, Antonescu CR, Charville GW, Sangoi AR. MEIS1::NCOA1 Primitive Spindle Cell Sarcoma of the Kidney : Report of 7 Cases of a Distinctive Clinicopathologic Entity. Am J Surg Pathol 2025; 49:620-632. [PMID: 40160183 DOI: 10.1097/pas.0000000000002386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Primitive sarcomas harboring the MEIS1::NCOA2 gene fusion were originally described in the kidney in 2018, and subsequently reported in other organs. These variably cellular neoplasms feature monomorphic primitive plump spindle cells forming nodules and whorls in addition to nondescript fascicular, solid, and storiform patterns. They lack skeletal muscle differentiation in contrast to the primarily intraosseous rhabdomyosarcomas that harbor the same gene fusion. We describe 7 new primary primitive renal sarcomas with MEIS1::NCOA1 gene fusions. Although their morphology overlaps with that described in MEIS1::NCOA2 renal sarcoma, 3 of the 7 cases contained adipose tissue. The majority had intimately admixed entrapped cystic epithelial elements and demonstrated patchy immunoreactivity for estrogen receptor and nuclear labeling for WT1 protein, leading to the differential diagnosis of malignant mixed epithelial stromal tumor (MEST) in 4 cases and metanephric stromal tumor in one. The neoplasms demonstrate a broad spectrum of clinicopathologic features ranging from a bland low-grade neoplasm that metastasized 9 years after diagnosis to a high-grade sarcoma with multiple recurrences, ultimately leading to patient death in under 1 year. In summary, MEIS1::NCOA1 primitive sarcomas overlap with the previously described MEIS1::NCOA2 primitive renal sarcomas and represent a distinctive renal neoplasm that can be mistaken for malignant MEST. Grade ranges from low to high but even low-grade neoplasms require long-term clinical follow-up.
Collapse
Affiliation(s)
- Pedram Argani
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sintawat Wangsiricharoen
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | - Maria Tretiakova
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA
| | - Yajuan J Liu
- Department of Pathology and Laboratory Medicine, University of Washington, Seattle, WA
| | - Sara M Falzarano
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | | | - Fadi Brimo
- McGill University Health Center, Montreal, QC, Canada
| | - John M Gross
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ezra Baraban
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andres Matoso
- Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kristina Wakeman
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | - Christopher Corless
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | - Tanaya Neff
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | - Benjamin F Smith
- Department of Pathology and Laboratory Medicine, Oregon Health & Science University, Portland, OR
| | | | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| | - Cristina R Antonescu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Ankur R Sangoi
- Department of Pathology, Stanford University, Stanford, CA
| |
Collapse
|
2
|
Caliò A, Marletta S, Brunelli M, Antonini P, Martelli FM, Marcolini L, Stefanizzi L, Martignoni G. TFE3-Rearranged Tumors of the Kidney: An Emerging Conundrum. Cancers (Basel) 2024; 16:3396. [PMID: 39410016 PMCID: PMC11475521 DOI: 10.3390/cancers16193396] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Identical translocations involving the TFE3 gene and various partners have been found in both renal and soft tissue tumors, like alveolar soft part sarcoma (ASPSCR1), ossifying fibromyxoid tumor (PHF1), epithelioid hemangioendothelioma, and the clear cell stromal tumor of the lung (YAP1). Methods: Herein, we review in detail the clinicopathologic and molecular data of TFE3-rearranged renal tumors and propose our perspective, which may shed light on this emerging conundrum. Results: Among the kidney tumors carrying TFE3 translocations, most are morphologically heterogeneous carcinomas labeling for the tubular marker PAX8. The others are mesenchymal neoplasms known as PEComas, characterized by epithelioid cells co-expressing smooth muscle actin, cathepsin-K, melanogenesis markers, and sometimes melanin pigment deposition. Over the past 30 years, numerous TFE3 fusion partners have been identified, with ASPL/ASPSCR1, PRCC, SFPQ/PSF, and NONO being the most frequent. Conclusions: It is not well understood why similar gene fusions can give rise to renal tumors with different morpho-immunophenotypes, which may contribute to the recent disagreement regarding their classification. However, as these two entities, respectively, epithelial and mesenchymal in nature, are widely recognized by the pathology community and their clinicopathologic features well established, we overall believe it is still better to retain the names TFE3-rearranged renal cell carcinoma and TFE3-rearranged PEComa.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
| | - Stefano Marletta
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
- Division of Pathology, Humanitas Istituto Clinico Catanese, 95045 Catania, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
| | - Pietro Antonini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
| | - Filippo Maria Martelli
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
| | - Lisa Marcolini
- Department of Pathology, Pederzoli Hospital, 37019 Peschiera del Garda, Italy; (L.M.); (L.S.)
| | - Lavinia Stefanizzi
- Department of Pathology, Pederzoli Hospital, 37019 Peschiera del Garda, Italy; (L.M.); (L.S.)
| | - Guido Martignoni
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (A.C.); (S.M.); (M.B.); (P.A.); (F.M.M.)
- Department of Pathology, Pederzoli Hospital, 37019 Peschiera del Garda, Italy; (L.M.); (L.S.)
| |
Collapse
|
3
|
Wang Y, Guo X, Meng Z, Cui Y, Sun Y. Pregnant patient with Xp11.2/transcription factor E3 translocation renal cell carcinoma: a case report and literature review. Front Oncol 2024; 14:1388880. [PMID: 38988705 PMCID: PMC11233431 DOI: 10.3389/fonc.2024.1388880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
MiT family translocation renal cell carcinomas (tRCCs) primarily include Xp11.2/transcription factor E3 (TFE3) gene fusion-associated renal cell carcinoma (Xp11.2 tRCC) and t(6;11)/TFEB gene fusion-associated RCC. Clinical cases of these carcinomas are rare. Fluorescence in situ hybridization can be used to identify the type, but there are no standard diagnostic and treatment methods available, and the prognosis remains controversial. Herein, we present a case of a patient with Xp11.2 tRCC at 29 weeks of gestation. The baby was successfully delivered, and radical surgery was performed for renal cancer at the same time. This is a unique and extremely rare case. We have described the case and performed a literature review to report the progress of current research on the treatment and prognosis of pregnant patients with Xp11.2/TFE3 translocation renal cell carcinoma. This study aims to contribute to improving the diagnosis and treatment of Xp11.2 tRCC in pregnant patients.
Collapse
Affiliation(s)
- Yanchen Wang
- Department of Urology, Weifang People's Hospital, Weifang, Shandong, China
| | - Xiaoyan Guo
- Department of Nuclear Medicine, Weifang People'sHospital, Weifang, Shandong, China
| | - Zhe Meng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yong Cui
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yaofei Sun
- Department of Urology, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
4
|
Zhang X, Bolck HA, Rupp NJ, Moch H. Genomic alterations and diagnosis of renal cancer. Virchows Arch 2024; 484:323-337. [PMID: 37999735 PMCID: PMC10948545 DOI: 10.1007/s00428-023-03700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/24/2023] [Accepted: 11/04/2023] [Indexed: 11/25/2023]
Abstract
The application of molecular profiling has made substantial impact on the classification of urogenital tumors. Therefore, the 2022 World Health Organization incorporated the concept of molecularly defined renal tumor entities into its classification, including succinate dehydrogenase-deficient renal cell carcinoma (RCC), FH-deficient RCC, TFE3-rearranged RCC, TFEB-altered RCC, ALK-rearranged RCC, ELOC-mutated RCC, and renal medullary RCC, which are characterized by SMARCB1-deficiency. This review aims to provide an overview of the most important molecular alterations in renal cancer, with a specific focus on the diagnostic value of characteristic genomic aberrations, their chromosomal localization, and associations with renal tumor subtypes. It may not yet be the time to completely shift to a molecular RCC classification, but undoubtedly, the application of molecular profiling will enhance the accuracy of renal cancer diagnosis, and ultimately guide personalized treatment strategies for patients.
Collapse
Affiliation(s)
- Xingming Zhang
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Hella A Bolck
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstr. 12, 8091, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Chen X, Wang W, Yeh J, Wu Y, Oehler VG, Naresh KN, Liu YJ. Clinical Validation of FusionPlex RNA Sequencing and Its Utility in the Diagnosis and Classification of Hematologic Neoplasms. J Mol Diagn 2023; 25:932-944. [PMID: 37813298 DOI: 10.1016/j.jmoldx.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 08/11/2023] [Accepted: 09/12/2023] [Indexed: 10/11/2023] Open
Abstract
Recurrent gene rearrangements result in gene fusions that encode chimeric proteins, driving the pathogenesis of many hematologic neoplasms. The fifth edition World Health Organization classification and International Consensus Classification 2022 include an expanding list of entities defined by such gene rearrangements. Therefore, sensitive and rapid methods are needed to identify a broad range of gene fusions for precise diagnosis and prognostication. In this study, we validated the FusionPlex Pan-Heme panel analysis using anchored multiplex PCR/targeted RNA next-generation sequencing for routine clinical testing. Furthermore, we assessed its utility in detecting gene fusions in myeloid and lymphoid neoplasms. The validation cohort of 61 cases demonstrated good concordance between the FusionPlex Pan-Heme panel and other methods, including chromosome analysis, fluorescence in situ hybridization, RT-PCR, and Sanger sequencing, with an analytic sensitivity and specificity of 95% and 100%, respectively. In an independent cohort of 28 patients indicated for FusionPlex testing, gene fusions were detected in 21 patients. The FusionPlex Pan-Heme panel analysis reliably detected fusion partners and patient-specific fusion sequences, allowing accurate classification of hematologic neoplasms and the discovery of new fusion partners, contributing to a better understanding of the pathogenesis of the diseases.
Collapse
Affiliation(s)
- Xueyan Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Wenjing Wang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Jeffrey Yeh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Yu Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Vivian G Oehler
- Department of Medicine, University of Washington, Seattle, Washington
| | - Kikkeri N Naresh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington.
| |
Collapse
|
6
|
Agaimy A, Perret R, Demicco EG, Gross J, Liu YJ, Azmani R, Engelmann C, Schubart C, Seppet J, Stoehr R, Le Loarer F, Dickson BC. GAB1::ABL1 fusions define a distinctive soft tissue neoplasm, with variable perineurial differentiation, and a predilection for children and young adults. Genes Chromosomes Cancer 2023; 62:449-459. [PMID: 36744864 DOI: 10.1002/gcc.23131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/14/2023] [Accepted: 02/01/2023] [Indexed: 02/07/2023] Open
Abstract
Although well known as a fusion partner in hematological malignancies, fusion genes involving the ABL proto-oncogene 1 (ABL1), mapping to chromosomal region 9q34.12, have only been anecdotally reported in five soft tissue tumors. These neoplasms have been variously reported as perineurioma, angiofibroma, and solitary fibrous tumor, and all have harbored a GAB1::ABL1 gene fusion; however, the nosology and clinicopathological characteristics of soft tissue tumors carrying this rare fusion have not been delineated. We herein describe eight tumors containing the GAB1::ABL1 fusion and review previously reported cases in a series to define their morphological spectrum, address immunohistochemical evidence for a line of differentiation, with special reference to the presence or absence of a perineurial immunophenotype, and gather insight into their behavior. The patients included four females and four males, aged 13-37 years (median, 24 years). Two cases each originated in the shoulder area, trunk, hands, and lower extremities, with a size range of 1.5-8 cm (median, 3.4 cm). Four tumors were deep and four superficial. All tumors were morphologically similar, being composed of bland fibroblast-like spindle to ovoid cells diffusely arranged in a paucivascular fibrous to fibromyxoid stroma with variable resemblance to soft tissue perineurioma. Mitotic activity was generally low (0-8 mitoses in 10 high-power fields [HPFs]; median, 1). All lesions had at least focally infiltrative margins, but they otherwise lacked pleomorphism and necrosis. Immunohistochemistry showed focal reactivity for CD34 (5/7), epithelial membrane antigen (EMA) (3/8), claudin1 (2/3), GLUT1 (4/6), and S100 (2/7); other markers, including MUC4 (0/7), desmin (0/9), and smooth muscle actin (SMA) (0/4), were negative. RNA sequencing revealed a GAB1::ABL1 fusion in all cases with exon 6 of GAB1 fused to exon 2 of ABL1. Treatments included various forms of surgical intervention in seven cases; one tumor was biopsied only. Limited follow-up was available for five patients. One tumor regrew rapidly within 1 month to 1.5 cm after an initial marginal excision and was re-excised with close margins. Four patients were disease-free at 1, 3, 14, and 25 months of follow-up. Metastases have not, to date, been observed. This series characterizes "GAB1::ABL1 fusion-positive spindle cell neoplasm" as a distinct entity, with overlapping features with soft tissue perineurioma and predilection for children and young adults.
Collapse
Affiliation(s)
- Abbas Agaimy
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center, European Metropolitan Area Erlangen-Nuremberg (CCC ER-EMN), Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Raul Perret
- Department of Biopathology, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
| | - Elizabeth G Demicco
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - John Gross
- Department of Pathology, The Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, Clinical Genomics Laboratory, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rihab Azmani
- Bioinformatics, Data and Digital Health Department, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
| | - Carsten Engelmann
- Department of Pediatric Surgery, Brandenburg Clinic Center, Berlin, Germany
| | - Christoph Schubart
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center, European Metropolitan Area Erlangen-Nuremberg (CCC ER-EMN), Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - Joosep Seppet
- Pathology Department, Tartu University Hospital, Tartu, Estonia
| | - Robert Stoehr
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center, European Metropolitan Area Erlangen-Nuremberg (CCC ER-EMN), Friedrich Alexander University of Erlangen-Nuremberg, Erlangen, Germany
| | - François Le Loarer
- Department of Biopathology, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- Bordeaux Institute of Oncology, BRIC U1312, INSERM, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Brendan C Dickson
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Kallen ME, Michal M, Meyer A, Suster DI, Olson NJ, Charville GW, Perret R, Gross JM. Calcified Chondroid Mesenchymal Neoplasm: Exploring the Morphologic and Clinical Features of an Emergent Entity With a Series of 33 Cases. Am J Surg Pathol 2023; 47:725-737. [PMID: 37102574 DOI: 10.1097/pas.0000000000002044] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Calcified chondroid mesenchymal neoplasm is a term proposed for tumors with a spectrum of morphologic features, including cartilage/chondroid matrix formation, that frequently harbor FN1 gene fusions. We report a series of 33 cases of putative calcified chondroid mesenchymal neoplasms, mostly referred for expert consultation out of concern for malignancy. Patients included 17 males and 16 females, with a mean age of 51.3 years. Anatomic locations include the hands and fingers, feet and toes, head and neck, and temporomandibular joint; 1 patient presented with multifocal disease. Radiologic review showed soft tissue masses with variable internal calcification, which occasionally scalloped bone but in all cases appeared indolent/benign. Tumors had a mean gross size of 2.1 cm and a homogenous rubbery to fibrous/gritty tan-white cut surface. Histology demonstrated multinodular architecture with a prominent chondroid matrix and increased cellularity towards the periphery of the nodules. The tumor cells were polygonal with eccentric nuclei and bland cytologic features and showed a variable amount of increased spindled / fibroblastic forms in the perinodular septa. The majority of cases had notable grungy and/or lacy calcifications. A subset of cases demonstrated at least focal areas of increased cellularity and osteoclast-like giant cells. Herein, we confirm the distinct morphologic and clinicopathologic features associated with this entity with the largest series to date, with a focus on practical diagnostic separation from similar chondroid neoplasms. Awareness of these features is critical in avoiding pitfalls, including a malignant diagnosis of chondrosarcoma.
Collapse
Affiliation(s)
- Michael E Kallen
- Department of Pathology, University of Maryland School of Medicine
| | | | - Anders Meyer
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, KS
| | - David I Suster
- Department of Pathology, Immunology, and Laboratory Medicine, Rutgers New Jersey Medical School, Rutgers, NJ
| | - Nicholas J Olson
- Physicians Laboratory
- University of South Dakota Sanford School of Medicine, Sioux Falls, SD
| | | | - Raul Perret
- Institut Bergonié / Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - John M Gross
- Department of Pathology, Johns Hopkins Medicine, Baltimore, MD
| |
Collapse
|
8
|
Lin J, Tang Z, Zhang C, Dong W, Liu Y, Huang H, Liu H, Huang J, Lin T, Chen X. TFE3 gene rearrangement and protein expression contribute to a poor prognosis of renal cell carcinoma. Heliyon 2023; 9:e16076. [PMID: 37215783 PMCID: PMC10196445 DOI: 10.1016/j.heliyon.2023.e16076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/30/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Background TFE3-rearranged renal cell carcinoma (TFE3-rearranged RCC) is a type of kidney cancer with a low incidence, with no consensus about whether it has a worse prognosis than clear cell renal cell carcinoma (ccRCC). This study attempted to elucidate the impact of TFE3-rearranged RCC by analyzing its clinical features and prognosis. Methods Patients treated in Sun Yat-sen Memorial Hospital (SYSMH) who were suspected to be diagnosed with TFE3-rearranged RCC were divided into two groups, TFE3-rearranged RCC and ccRCC with positive TFE3 protein expression on immunohistochemistry [TFE3(+) ccRCC], by dual-color, break-apart fluorescence in situ hybridization (FISH). After balancing the baseline characteristics with TFE3(+) ccRCC using the propensity score matching (PSM) method in a ratio of 2, we selected patients diagnosed with ccRCC with negative TFE3 protein expression on immunohistochemistry [TFE3(-) ccRCC]. The impact of TFE3 gene rearrangement and protein expression on renal cell carcinoma was determined by feature comparison with a nonparametric test and survival analysis with the Kaplan‒Meier method. Results Among 37 patients suspected of having TFE3-rearranged RCC, 13 patients were diagnosed with TFE3-rearranged RCC, and 24 patients had TFE3(+) ccRCC. The recurrence and new metastasis of TFE3-rearranged RCC was relatively common, even if the tumor stage was early at the first diagnosis. Through feature comparison and survival analysis, we found that TFE3-rearranged RCC was quite similar to TFE3(+) ccRCC. Compared with TFE3(-) ccRCC, TFE3(+) ccRCC tended to have a larger tumor diameter (P = 0.011), higher neutrophil/lymphocyte ratio (NLR) (P = 0.017) and metastatic potential (P = 0.022), and worse overall survival (OS) (P = 0.043) and PFS (P = 0.016). The survival analysis showed that TFE3-rearranged RCC had a worse PFS than ccRCC (P = 0.002), and TFE3(+) RCC had a worse PFS than TFE3(-) RCC (P = 0.001). According to the stratification system based on the combination of TFE3 and lymphovascular invasion (LVI), we further found that the prognosis from good to poor was TFE3(-) LVI(-), TFE3(+) LVI(-), TFE3(+) LVI(+) and TFE3(-) LVI(+), with statistically significant differences in both OS (P = 0.001) and PFS (P < 0.001). In addition, we also reported two cases with poor prognosis, of which one was TFE3-rearranged RCC and the other was TFE3(+) ccRCC. Conclusions This is a novel finding that both FISH confirmed TFE3 gene rearrangement-mediated TFE3-rearranged RCC and IHC confirmed positive TFE3 protein expression [TFE3(+)] contribute to a poor prognosis in RCC, suggesting more active treatment and careful follow-up for TFE3(+) RCC patients. The combination of TFE3 and LVI may be a new risk stratification system for RCC.
Collapse
Affiliation(s)
- Junyi Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, Guangzhou Urology Research Institute, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhuang Tang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Chengjunyu Zhang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Wen Dong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Yeqing Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120, China
| | - Hao Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Hao Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| | - Xu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, 510120, China
| |
Collapse
|
9
|
Gandhi J, Mantilla JG, Ricciotti RW, Chen EY, Liu YJ, Bandhlish A. Myoepithelial carcinoma of the parotid gland with a novel CTCF::NCOA2 fusion. Genes Chromosomes Cancer 2023; 62:161-166. [PMID: 36331420 DOI: 10.1002/gcc.23101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 10/29/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
We describe a case of a myoepithelial carcinoma of the superficial parotid gland in a 46-year-old male harboring a novel CTCF::NCOA2 gene fusion. To our knowledge, this novel gene fusion has not been described previously in myoepithelial carcinoma. A 46-year-old male patient presented with a mass involving the superficial left parotid gland with extension into the external auditory canal (EAC) and erosion of the conchal cartilage. Histologically, the neoplasm was composed of uniform spindled, epithelioid/ovoid cells arranged in cords and nests within hyalinized to myxoid stroma. On immunohistochemistry (IHC), the tumor cells demonstrated patchy and variable staining for low molecular weight cytokeratin (CAM5.2), pan-cytokeratin (OSCAR), and S-100. Overall, the morphological and immunohistochemical attributes supported a locally aggressive tumor of myoepithelial differentiation consistent with myoepithelial carcinoma. Molecular analysis using a custom 115-gene gene panel by targeted RNA sequencing, showed an in-frame CTCF::NCOA2 fusion. In addition to reporting this novel fusion in myoepithelial carcinoma, we also discuss relevant differential diagnosis, and provide a brief review of NCOA2 gene function in both normal and neoplastic contexts.
Collapse
Affiliation(s)
- Jatin Gandhi
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA.,Department of Laboratory Medicine and Pathology, Emory University School of Medicine, Atlanta, Washington, USA
| | - Jose G Mantilla
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Robert W Ricciotti
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Anshu Bandhlish
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
10
|
Miller TI, Mantilla JG, Wang W, Liu YJ, Tretiakova M. Novel low-grade renal spindle cell neoplasm with HEY1::NCOA2 fusion that is distinct from mesenchymal chondrosarcoma. Genes Chromosomes Cancer 2023; 62:171-175. [PMID: 36416671 DOI: 10.1002/gcc.23105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
HEY1-NCOA2 fusion is most described in mesenchymal chondrosarcoma. This is the first case report of a primary renal spindle cell neoplasm of uncertain malignant potential with a HEY1::NCOA2 fusion identified by Fusionplex RNA-sequencing that is histologically distinct from mesenchymal chondrosarcoma. The neoplasm was identified in a 33-year-old woman without significant past medical history who underwent partial nephrectomy for an incidentally discovered renal mass. The histologic features of the mass included spindle cells with variable cellularity and monotonous bland cytology forming vague fascicles and storiform architecture within a myxoedematous and collagenous stroma with areas of calcification. The morphologic and immunophenotypic features were not specific for any entity but were most similar to low-grade fibromyxoid sarcoma. To date, the patient has not had recurrence, and the malignant potential of the neoplasm is uncertain.
Collapse
Affiliation(s)
- Timothy Isaac Miller
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jose G Mantilla
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Wenjing Wang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Paulson VA, Liu YJ, Fang H, Browd SR, Hauptman JS, Wright J, Lockwood CM, Leary SES, Cole BL. Infantile ZFTA Fusion-Positive Tumor of the Posterior Fossa: Molecular Tumor Board. JCO Precis Oncol 2023; 7:e2200226. [PMID: 36862968 DOI: 10.1200/po.22.00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Affiliation(s)
- Vera A Paulson
- Department of Laboratory Medicine and Pathology, Genetics and Solid Tumor Laboratory, University of Washington, Seattle, WA.,Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, Clinical Genomics Laboratory, University of Washington School of Medicine, Seattle, WA.,Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - He Fang
- Department of Laboratory Medicine and Pathology, Clinical Genomics Laboratory, University of Washington School of Medicine, Seattle, WA
| | - Sam R Browd
- Division of Neurosurgery, Department of Neurological Surgery, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - Jason S Hauptman
- Division of Neurosurgery, Department of Neurological Surgery, University of Washington, Seattle Children's Hospital, Seattle, WA
| | - Jason Wright
- Radiology, Seattle Children's Hospital, University of Washington; Seattle, WA
| | - Christina M Lockwood
- Department of Laboratory Medicine and Pathology, Genetics and Solid Tumor Laboratory, University of Washington, Seattle, WA.,Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Sarah E S Leary
- Cancer and Blood Disorders Center, Seattle Children's Hospital; Department of Pediatrics, University of Washington; Center for Clinical and Translational Research, Seattle Children's Research Institute, Seattle, WA.,Fred Hutchinson Cancer Research Center, Seattle, WA.,Brotman Baty Institute for Precision Medicine, Seattle, WA
| | - Bonnie L Cole
- Department of Laboratory Medicine and Pathology, Clinical Genomics Laboratory, University of Washington School of Medicine, Seattle, WA.,Department of Laboratories, Seattle Children's Hospital, University of Washington; Seattle, WA.,Brotman Baty Institute for Precision Medicine, Seattle, WA
| |
Collapse
|
12
|
Rottmann D, Abdulfatah E, Pantanowitz L. Molecular testing of soft tissue tumors. Diagn Cytopathol 2023; 51:12-25. [PMID: 35808975 PMCID: PMC10084007 DOI: 10.1002/dc.25013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The diagnosis of soft tissue tumors is challenging, especially when the evaluable material procured is limited. As a result, diagnostic ancillary testing is frequently needed. Moreover, there is a trend in soft tissue pathology toward increasing use of molecular results for tumor classification and prognostication. Hence, diagnosing newer tumor entities such as CIC-rearranged sarcoma explicitly requires molecular testing. Molecular testing can be accomplished by in situ hybridization, polymerase chain reaction, as well as next generation sequencing, and more recently such testing can even be accomplished leveraging an immunohistochemical proxy. CONCLUSION This review evaluates the role of different molecular tests in characterizing soft tissue tumors belonging to various cytomorphologic categories that have been sampled by small biopsy and cytologic techniques.
Collapse
Affiliation(s)
- Douglas Rottmann
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Eman Abdulfatah
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Liron Pantanowitz
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Maughan BL. Start of a New Era: Management of Non-Clear Cell Renal Cell Carcinoma in 2022. Curr Oncol Rep 2022; 24:1201-1208. [PMID: 35438388 PMCID: PMC9468090 DOI: 10.1007/s11912-022-01269-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW Historically, kidney cancer was diagnosed as either clear cell renal carcinoma (ccRCC) or non-clear cell renal carcinoma (nccRCC). With further research into the pathophysiology of nccRCC, multiple distinct subtypes have emerged creating distinct diagnosis, such as papillary renal cell carcinoma (PRCC), chromophobe renal cell carcinoma (crRCC), or unclassified carcinoma (cRCC). Many other kidney cancer subtypes are now included in the WHO classification system. RECENT FINDINGS The prognosis for each of the more frequently diagnosed types is discussed here along with treatment recommendations. The available clinical trial results and salient retrospective studies of each subtype are reviewed here to guide clinicians on the optimal treatment selection for patients with these rare histologic types or RCC. Many nccRCC types are now recognized and each has unique molecular drivers which are different than ccRCC. The optimal treatment strategy is different for each subtype. The prognosis also differs based on the histology.
Collapse
Affiliation(s)
- Benjamin L Maughan
- Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Room HCI S 5617, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
14
|
Simonaggio A, Ambrosetti D, Verkarre V, Auvray M, Oudard S, Vano YA. MiTF/TFE Translocation Renal Cell Carcinomas: From Clinical Entities to Molecular Insights. Int J Mol Sci 2022; 23:ijms23147649. [PMID: 35886994 PMCID: PMC9324307 DOI: 10.3390/ijms23147649] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 12/30/2022] Open
Abstract
MiTF/TFE translocation renal cell carcinoma (tRCC) is a rare and aggressive subtype of RCC representing the most prevalent RCC in the pediatric population (up to 40%) and making up 4% of all RCCs in adults. It is characterized by translocations involving either TFE3 (TFE3-tRCC), TFEB (TFEB-tRCC) or MITF, all members of the MIT family (microphthalmia-associated transcriptional factor). TFE3-tRCC was first recognized in the World Health Organization (WHO) classification of kidney cancers in 2004. In contrast to TFEB-tRCC, TFE3-tRCC is associated with many partners that can be detected by RNA or exome sequencing. Both diagnoses of TFE3 and TFEB-tRCC are performed on morphological and immunohistochemical features, but, to date, TFE break-apart fluorescent in situ hybridization (FISH) remains the gold standard for diagnosis. The clinical behavior of tRCC is heterogeneous and more aggressive in adults. Management of metastatic tRCC is challenging, especially in the younger population, and data are scarce. Efficacy of the standard of care-targeted therapies and immune checkpoint inhibitors remains low. Recent integrative exome and RNA sequencing analyses have provided a better understanding of the biological heterogeneity, which can contribute to a better therapeutic approach. We describe the clinico-pathological entities, the response to systemic therapy and the molecular features and techniques used to diagnose tRCC.
Collapse
Affiliation(s)
- Audrey Simonaggio
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP. Centre—Université Paris-Cité, F-75015 Paris, France; (A.S.); (M.A.); (S.O.)
| | - Damien Ambrosetti
- Department of Pathology, CHU Nice, Université Côte d’Azur, F-06107 Nice, France;
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR 7284, INSERM U1081, University Côte d’Azur, F-06107 Nice, France
| | - Virginie Verkarre
- Department of Pathology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP. Centre—Université Paris-Cité, F-75015 Paris, France;
- INSERM UMR-970, PARCC, Université Paris-Cité, F-75015 Paris, France
| | - Marie Auvray
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP. Centre—Université Paris-Cité, F-75015 Paris, France; (A.S.); (M.A.); (S.O.)
| | - Stéphane Oudard
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP. Centre—Université Paris-Cité, F-75015 Paris, France; (A.S.); (M.A.); (S.O.)
- INSERM UMR-970, PARCC, Université Paris-Cité, F-75015 Paris, France
| | - Yann-Alexandre Vano
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP. Centre—Université Paris-Cité, F-75015 Paris, France; (A.S.); (M.A.); (S.O.)
- INSERM UMR-970, PARCC, Université Paris-Cité, F-75015 Paris, France
- Centre de Recherche des Cordeliers, INSERM, Université Paris-Cité, Sorbonne Université, F-75006 Paris, France
- Correspondence: ; Tel.: +33-624281311
| |
Collapse
|
15
|
Tretiakova MS. Chameleon TFE3-translocation RCC and How Gene Partners Can Change Morphology: Accurate Diagnosis Using Contemporary Modalities. Adv Anat Pathol 2022; 29:131-140. [PMID: 35180736 DOI: 10.1097/pap.0000000000000332] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Translocation renal cell carcinoma (tRCC) with TFE3 gene rearrangements has been born as a distinct entity 20 years ago. These relatively rare tumors were notable among other RCC subtypes because of their disproportionally high incidence among children and young adults. Initial reports were focused on describing unifying morphologic criteria and typical clinical presentation. Follow-up studies of ancillary immunohistochemical and hybridization techniques provided additional diagnostic tools allowing recognition of tRCC tumors in practice. However, a growing body of literature also expanded the clinicomorphologic spectrum of tRCCs, to include a significant morphologic overlap with other RCC variants thus blurring the diagnostic clarity of this entity. More recent molecular studies utilizing next-generation sequencing technology accelerated recognition of numerous novel gene partners fusing at different breakpoints with the TFE3 gene. Accumulating data indicates that morphologic and clinical heterogeneity of tRCC could be explained by fusion subtypes, and knowledge of TFE3 partnering genes may be important in predicting tumor behavior. Herein we provided a comprehensive analysis of ∼400 tRCC cases with known TFE3 fusion partners, estimated their relative incidence and summarized clinicomorphologic features associated with most common fusion subtypes. Our data was based on an extensive literature review and had a special focus on comparing immunohistochemistry, fluorescent in situ hybridization and contemporary molecular studies for the accurate diagnosis of tRCC.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA
| |
Collapse
|
16
|
Ge Y, Lin X, Zhang Q, Lin D, Luo L, Wang H, Li Z. Xp11.2 Translocation Renal Cell Carcinoma With TFE3 Rearrangement: Distinct Morphological Features and Prognosis With Different Fusion Partners. Front Oncol 2021; 11:784993. [PMID: 34917511 PMCID: PMC8668609 DOI: 10.3389/fonc.2021.784993] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/08/2021] [Indexed: 01/20/2023] Open
Abstract
BackgroundRenal cell carcinoma (RCC) associated with Xp11.2 translocation/TFE3 gene fusion is a rare and new subtype of RCC and was classified by the WHO in 2004. Since then, multiple 5′ fusion partners for TFE3 have been reported; however, the impact of individual fusion variant on specific clinicopathologic features of Xp11.2 RCCs has not been well defined.MethodsFour Xp11.2 translocation RCCs were identified by morphological, immunostaining, and fluorescence in situ hybridization (FISH) assays from 200 patients who attended Guangdong General Hospital between January 2017 and January 2020. All these four cases were further analyzed by RNA sequencing to explore their TFE3 gene fusion partners. The clinicopathologic features, including clinical manifestations, pathological findings, treatment strategies, clinical outcomes, and follow-up information on Xp11.2 translocation RCCs, were recorded and evaluated.ResultsThese four cases affected one male and three females. The median age was 13 years at the time of diagnosis (range = 4–20 years). All the examined tumors were unilateral and unifocal. The largest diameter of these tumors ranged from 2.0 to 10.0 cm, and the average was 5.55 cm. Regional lymph node or distant metastasis developed in two patients. Three cases demonstrated known fusions: ASPCR1–TFE3 (two cases) and PRCC–TFE3 (one case). However, one case showed an unreported VCP–TFE3 fusion gene in Xp11.2 translocation RCCs. Immunohistochemistry results revealed tumor cells diffusely positive for TFE3, but have no consistency in other markers. Moreover, there were different clinical prognoses among the different variant TFE3 rearrangements; RCC patients with VCP–TFE3 translocation had worse prognosis compared to those with other fusion types. Follow-up were available for all the patients and ranged from 3 to 36 months. Three patients were without evidence of disease progression, while that with VCP–TFE3 fusion died of the disease 3 months after the diagnosis.ConclusionIn conclusion, our data expand the list of TFE3 gene fusion partners and the clinicopathologic features of Xp11.2 RCCs with specific TFE3 gene fusions. We identified a novel VCP–TFE3 fusion in Xp11.2 translocation RCCs for the first time, which has unique morphology and worse prognosis than those with other variant TFE3 rearrangements. Integration of morphological, immunohistochemical, and molecular methods is often necessary for the precise diagnosis and optimal clinical management of malignant tumors.
Collapse
Affiliation(s)
- Yan Ge
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xingtao Lin
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Danyi Lin
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Luqiao Luo
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huiling Wang
- Department of General Surgery, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Huiling Wang, ; Zhi Li,
| | - Zhi Li
- Department of Pathology, Guangdong Provincial People’s Hospital/Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Huiling Wang, ; Zhi Li,
| |
Collapse
|
17
|
Tan SY, Al-Ibraheemi A, Ahrens WA, Oesterheld JE, Fanburg-Smith JC, Liu YJ, Spunt SL, Rudzinski ER, Coffin C, Davis JL. ALK rearrangements in infantile fibrosarcoma-like spindle cell tumours of soft tissue and kidney. Histopathology 2021; 80:698-707. [PMID: 34843129 DOI: 10.1111/his.14603] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022]
Abstract
AIMS Recurrent alterations in receptor tyrosine kinase (RTK) and downstream effectors are described in infantile fibrosarcoma (IFS)/cellular congenital mesoblastic nephroma (cCMN) and a subset of spindle cell sarcomas, provisionally designated 'NTRK-rearranged' spindle cell neoplasms. These two groups of tumours demonstrate overlapping morphologies and harbour alterations in NTRK1/2/3, RET, MET, ABL1, ROS1, RAF1 and BRAF, although their relationship is not fully elucidated. We describe herein a cohort of paediatric tumours with clinicopathological features not typical for inflammatory myofibroblastic tumour, but rather with similarities to cCMN/IFS harbouring ALK fusions. METHODS AND RESULTS Clinicopathological features were assessed and partner agnostic targeted RNA sequencing on clinically validated platforms were performed. Tumours occurred in patients aged from 2 to 10 years (median age 2 years) with a 2:2 male to female ratio and an average size of 8.4 cm. Two tumours arose in soft tissues and two in the kidney. Morphological features included spindle to ovoid cells arranged in long fascicles or haphazardly within a myxoid to collagenised stroma; a subset of cases had either dilated, ectatic vessels or focal perivascular hyalinosis. By immunohistochemistry, all cases tested showed cytoplasmic expression of anaplastic lymphoma kinase (ALK) and one case demonstrated co-expression of CD34 and S100. CONCLUSIONS This series of ALK-rearranged IFS-like tumours expands the spectrum of targetable kinases altered in these tumours and reinforces the potential overlap between IFS/cCMN-like tumours and the provisional entity of 'NTRK-rearranged' spindle cell neoplasms.
Collapse
Affiliation(s)
- Serena Y Tan
- Department of Pathology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alyaa Al-Ibraheemi
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Javier E Oesterheld
- Department of Pediatrics, Levine Children's Hospital, Atrium Health, Charlotte, NC, USA
| | - Julie C Fanburg-Smith
- Department of Pathology, Pediatrics and Orthopedics, PennState Health, Penn State Children's Hospital, Penn State College of Medicine, Hershey, PA, USA
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Sheri L Spunt
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Erin R Rudzinski
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA
| | - Cheryl Coffin
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Jessica L Davis
- Department of Pathology, Oregon Health & Sciences University, Portland, OR, USA
| |
Collapse
|
18
|
Shi Q, Liu N, Yang L, Chen Y, Lu Y, Guo H, Han X, Li D, Gan W. Estradiol increases risk of topoisomerase IIβ-mediated DNA strand breaks to initiate Xp11.2 translocation renal cell carcinoma. Cell Commun Signal 2021; 19:114. [PMID: 34784933 PMCID: PMC8594210 DOI: 10.1186/s12964-021-00790-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Xp11.2 translocation renal cell carcinoma (tRCC) is defined by translocation of the transcription factor E3 (TFE3) gene located on chromosome Xp11.2. Due to the high incidence in women, 17β-estradiol (E2) may be a factor influencing TFE3 breaks, and the topoisomerase II (TOP2) poison is considered one of the important risk factors in mediating DNA breaks. In this study, we investigated the potential pathogenesis of Xp11.2 tRCC using the renal epithelial cell line HK-2. METHODS Immunofluorescence assay was performed to analyze DNA breaks by quantifying phosphorylation of H2AX (γH2AX), and the micronuclei (MN) assay was designed for monitoring chromosome breaks. The chromatin immunoprecipitation (CHIP) was used to detect whether proteins bound to specific DNA site, and the co-immunoprecipitation (Co-IP) was used to confirm whether proteins bound to other proteins. In some experiments, siRNA and shRNA were transfected to knockdown target genes. RESULTS Our results demonstrated that DNA double-strand breaks were mediated by TOP2β in HK-2 cells, and this process could be amplified through estrogen receptor α (ERα)-dependent pathway induced by E2. After performing translocation site analysis using target region sequencing data in two Xp11.2 tRCC cell lines and ten Xp11.2 tRCC patients, we confirmed that TOP2β and ERα could both bind to TFE3 translocation sites directly to mediate DNA breaks in a E2-dependent manner. However, TOP2β and ERα were not observed to have direct interaction, indicating that their collaborative may be implemented in other ways. Besides, TFE3 was found to be upregulated through NRF1 with increasing E2 concentration, which could increase the risk of TFE3 breaks. CONCLUSION Our results indicate that E2 amplifies TOP2β-mediated TFE3 breaks by ERα-dependent pathway, and E2 upregulates TFE3 by NRF1 to increase the risk of TFE3 breaks. This suggests that E2 is an important pathogenic factor for Xp11.2 tRCC pathogenesis. Video Abstract.
Collapse
Affiliation(s)
- Qiancheng Shi
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu China
| | - Ning Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu China
| | - Lei Yang
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu China
| | - Yi Chen
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu China
| | - Yanwen Lu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu China
| | - Xiaodong Han
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu China
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory and State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu China
| | - Weidong Gan
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu China
| |
Collapse
|
19
|
Brady C, Manning SC, Rudzinski E, Paulson V, Wang X, Liu YJ, Parikh SR, Bonilla-Velez J, Hawkins DS, Dahl J. Clinical Outcomes of Diffuse Sclerosing Variant Papillary Thyroid Carcinoma in Pediatric Patients. Laryngoscope 2021; 132:1132-1138. [PMID: 34713899 DOI: 10.1002/lary.29926] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/28/2021] [Accepted: 10/15/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVES/HYPOTHESIS The diffuse sclerosing variant of papillary thyroid carcinoma (DSV) may be more aggressive than conventional well-differentiated non-DSV related papillary thyroid carcinomas (N-PTC). STUDY DESIGN Retrospective chart review. METHODS Retrospective review of clinical outcomes for patients 21 years of age or younger who underwent initial surgery for PTC at a single institution from January 1, 2005 to April 1, 2020. Genomic analysis was performed using targeted next-generation sequencing. Data were analyzed using Fischer's exact test and Kaplan-Meier curve log-rank test. RESULTS Our cohort consisted of 72 patients, nine with DSV and 63 with N-PTC. Age at diagnosis was comparable (15.4 vs. 16.2 years, respectively, P = .46). DSV were more likely to be in the high-risk American Thyroid Academy pediatric risk group (100% vs. 41.3%, P = .004), to present with regional cervical lymph node metastases (100% vs. 60.3%, P = .036), and to present with distant metastases (67% vs. 22%, P = .005). No mortality seen in either group over 27.5 (interquartile range 14.8, 46.00) months average follow-up. Throughout the follow-up period, DSV were more likely to experience progression than N-PTC (hazard ratio = 5.7 [95% confidence interval 1.7-20.0; P = .0056]). In a subset of 19 patients with aggressive disease who had molecular testing as part of clinical care we detected RET fusions in nearly all DSV compared to a minority of N-PTC (83% vs. 15.4%, P = .0095). CONCLUSIONS Pediatric patients with DSV have more advanced disease at diagnosis and are more likely to experience progression of disease compared to patients with N-PTC. The prevalence of RET fusions in our cohort recapitulates the frequency of this alteration described in prior studies. LEVEL OF EVIDENCE 4 Laryngoscope, 2021.
Collapse
Affiliation(s)
- Charles Brady
- UW Medicine, University of Washington School of Medicine, Seattle, Washington, U.S.A
| | - Scott C Manning
- Division of Pediatric Otolaryngology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Erin Rudzinski
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Vera Paulson
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Xing Wang
- Seattle Children's Research Division, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Yajuan J Liu
- Department of Laboratory Medicine and Pathology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Sanjay R Parikh
- Division of Pediatric Otolaryngology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Julianna Bonilla-Velez
- Division of Pediatric Otolaryngology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - Douglas S Hawkins
- Division of Pediatrics, Seattle Children's Hospital, Seattle, Washington, U.S.A
| | - John Dahl
- Division of Pediatric Otolaryngology, Seattle Children's Hospital, Seattle, Washington, U.S.A
| |
Collapse
|
20
|
Argani P. Translocation carcinomas of the kidney. Genes Chromosomes Cancer 2021; 61:219-227. [PMID: 34704642 DOI: 10.1002/gcc.23007] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/24/2021] [Indexed: 01/19/2023] Open
Abstract
The MiT subfamily of transcription factors includes TFE3, TFEB, TFEC, and MITF. Gene fusions involving two of these transcription factors have been well-characterized in renal cell carcinoma (RCC). The TFE3-rearranged RCC (also known as Xp11 translocation RCC) was first officially recognized in the 2004 World Health Organization (WHO) renal tumor classification. The TFEB-rearranged RCC, which typically harbor a t(6;11)(p21;q12) translocation which results in a MALAT1-TFEB gene fusion, were first officially recognized in the 2016 WHO renal tumor classification. These two subtypes of translocation RCC have many similarities. Both disproportionately involve young patients, although adult translocation RCC overall outnumber pediatric cases. Both often have unusual and distinctive morphologies; the TFE3-rearranged RCCs frequently have clear cells with papillary architecture and abundant psammoma bodies, while the TFEB-rearranged RCCs frequently have a biphasic appearance with both small and large epithelioid cells and nodules of basement membrane material. However, the morphology of these two neoplasms can overlap, with one mimicking the other or other more common renal neoplasms. Both of these RCC underexpress epithelial immunohistochemical markers, such as cytokeratin and epithelial membrane antigen, relative to most other RCC. Unlike other RCC, both frequently express the cysteine protease cathepsin k and often express melanocytic markers like HMB45 and Melan A. Finally, TFE3 and TFEB have overlapping functional activity as these two transcription factors frequently heterodimerize and bind to the same targets. Therefore, these two neoplasms are now grouped together under the heading of "MiT family translocation RCC." Approximately 50 renal cell carcinomas with gene fusions involving the anaplastic lymphoma kinase (ALK) gene have now been reported. While those with a Vinculin-ALK fusion have distinctive features (predilection to affect children with sickle cell trait and to show solid architecture with striking cytoplasmic vacuolization), other ALK-fusion RCCs have more varied clinical presentations and pathologic features. This review summarizes our current knowledge of these recently described RCC.
Collapse
Affiliation(s)
- Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Departments of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Reddy S, Hung LH, Sala-Torra O, Radich JP, Yeung CC, Yeung KY. A graphical, interactive and GPU-enabled workflow to process long-read sequencing data. BMC Genomics 2021; 22:626. [PMID: 34425749 PMCID: PMC8381503 DOI: 10.1186/s12864-021-07927-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Background Long-read sequencing has great promise in enabling portable, rapid molecular-assisted cancer diagnoses. A key challenge in democratizing long-read sequencing technology in the biomedical and clinical community is the lack of graphical bioinformatics software tools which can efficiently process the raw nanopore reads, support graphical output and interactive visualizations for interpretations of results. Another obstacle is that high performance software tools for long-read sequencing data analyses often leverage graphics processing units (GPU), which is challenging and time-consuming to configure, especially on the cloud. Results We present a graphical cloud-enabled workflow for fast, interactive analysis of nanopore sequencing data using GPUs. Users customize parameters, monitor execution and visualize results through an accessible graphical interface. The workflow and its components are completely containerized to ensure reproducibility and facilitate installation of the GPU-enabled software. We also provide an Amazon Machine Image (AMI) with all software and drivers pre-installed for GPU computing on the cloud. Most importantly, we demonstrate the potential of applying our software tools to reduce the turnaround time of cancer diagnostics by generating blood cancer (NB4, K562, ME1, 238 MV4;11) cell line Nanopore data using the Flongle adapter. We observe a 29x speedup and a 93x reduction in costs for the rate-limiting basecalling step in the analysis of blood cancer cell line data. Conclusions Our interactive and efficient software tools will make analyses of Nanopore data using GPU and cloud computing accessible to biomedical and clinical scientists, thus facilitating the adoption of cost effective, fast, portable and real-time long-read sequencing. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07927-1.
Collapse
Affiliation(s)
| | - Ling-Hong Hung
- School of Engineering and Technology, University of Washington, 98402, Tacoma, WA, USA
| | - Olga Sala-Torra
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 98109, Seattle, WA, USA
| | - Jerald P Radich
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 98109, Seattle, WA, USA.,Clinical Research Division, Kurt Enslein Endowed Chair, Fred Hutchinson Cancer Research Center, 98109, Seattle, WA, USA.,Department of Medicine, University of Washington, 98109, Seattle, WA, USA
| | - Cecilia Cs Yeung
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 98109, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, University of Washington, 98109, Seattle, WA, USA
| | - Ka Yee Yeung
- School of Engineering and Technology, University of Washington, 98402, Tacoma, WA, USA.
| |
Collapse
|
22
|
Liu YJ, Wang W, Yeh J, Wu Y, Mantilla JG, Fletcher CDM, Ricciotti RW, Chen EY. Calcified chondroid mesenchymal neoplasms with FN1-receptor tyrosine kinase gene fusions including FGFR2, FGFR1, MERTK, NTRK1, and TEK: a molecular and clinicopathologic analysis. Mod Pathol 2021; 34:1373-1383. [PMID: 33727696 DOI: 10.1038/s41379-021-00786-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 12/15/2022]
Abstract
Translocations involving FN1 have been described in a variety of neoplasms that share the presence of a cartilage matrix and may also contain a variable extent of calcification. Fusions of FN1 to FGFR1 or FGFR2 have been reported in nine soft tissue chondromas, mostly demonstrated indirectly by FISH analysis. Delineation of FN1 fusions with various partner genes will facilitate our understanding of the pathogenesis and diagnostic classification of these neoplasms. In this study, we present molecular, clinical, and pathologic features of 12 cartilaginous soft tissue neoplasms showing a predilection for the TMJ region and the distal extremities. We analyzed for gene fusions with precise breakpoints using targeted RNA-seq with a 115-gene panel. We detected gene fusions in ten cases, including three novel fusions, FN1-MERTK, FN1-NTRK1, and FN1-TEK, each in one case, recurrent FN1-FGFR2 fusion in five cases, FN1-FGFR1 in one case, and FGFR1-PLAG1 in one case. The breakpoints in the 5' partner gene FN1 ranged from exons 11-48, retaining the domains of a signal peptide, FN1, FN2, and/or FN3, while the 3' partner genes retained the transmembrane domain, tyrosine kinase (TK) domains, and/or Ig domain. The tumors are generally characterized by nodular/lobular growth of polygonal to stellate cells within a chondroid matrix, often accompanied by various patterns of calcification, resembling those described for the chondroblastoma-like variant of soft tissue chondroma. Additional histologic findings include extensive calcium pyrophosphate dihydrate deposition in two cases and features resembling tenosynovial giant cell tumor (TGCT). Overall, while the tumors from our series show significant morphologic overlap with chondroblastoma-like soft tissue chondroma, we describe findings that expand the morphologic spectrum of these neoplasms and therefore refer to them as "calcified chondroid mesenchymal neoplasms." These neoplasms represent a spectrum of chondroid/cartilage matrix-forming tumors harboring FN1-receptor TK fusions that include those classified as soft tissue chondroma as well as chondroid TGCT.
Collapse
Affiliation(s)
- Yajuan J Liu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.
| | - Wenjing Wang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Jeffrey Yeh
- Department of Biology, University of Washington, Seattle, WA, United States
| | - Yu Wu
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Jose G Mantilla
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Christopher D M Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Robert W Ricciotti
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
23
|
Kuroda N, Trpkov K, Gao Y, Tretiakova M, Liu YJ, Ulamec M, Takeuchi K, Agaimy A, Przybycin C, Magi-Galluzzi C, Fushimi S, Kojima F, Sibony M, Hang JF, Pan CC, Yilmaz A, Siadat F, Sugawara E, Just PA, Ptakova N, Hes O. ALK rearranged renal cell carcinoma (ALK-RCC): a multi-institutional study of twelve cases with identification of novel partner genes CLIP1, KIF5B and KIAA1217. Mod Pathol 2020; 33:2564-2579. [PMID: 32467651 DOI: 10.1038/s41379-020-0578-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 11/09/2022]
Abstract
ALK rearranged renal cell carcinoma (ALK-RCC) has recently been included in 2016 WHO classification as a provisional entity. In this study, we describe 12 ALK-RCCs from 8 institutions, with detailed clinical, pathological, immunohistochemical (IHC), fluorescence in situ hybridization (FISH), and next generation sequencing (NGS) analyses. Patients' age ranged from 25 to 68 years (mean, 46.3 years). Seven patients were females and five were males (M:F = 1:1.4). Tumor size ranged from 17 to 70 mm (mean 31.5, median 25 mm). The pTNM stage included: pT1a (n = 7), pT1b (n = 1), and pT3a (n = 4). Follow-up was available for 9/12 patients (range: 2 to 153 months; mean 37.6 months); 8 patients were alive without disease and one was alive with distant metastases. The tumors demonstrated heterogeneous, 'difficult to classify' morphology in 10/12 cases, typically showing diverse architectural and cellular morphologies, including papillary, tubular, tubulocystic, solid, sarcomatoid (spindle cell), rhabdoid, signet-ring cell, and intracytoplasmic vacuoles, often set in a mucinous background. Of the remaining two tumors, one showed morphology resembling mucinous tubular and spindle cell renal cell carcinoma (MTSC RCC-like) and one was indistinguishable from metanephric adenoma. One additional case also showed a focal metanephric adenoma-like area, in an otherwise heterogeneous tumor. By IHC, all tumors were diffusely positive for ALK and PAX8. In both cases with metanephric adenoma-like features, WT1 and ALK were coexpressed. ALK rearrangement was identified in 9/11 tumors by FISH. ALK fusion partners were identified by NGS in all 12 cases, including the previously reported: STRN (n = 3), TPM3 (n = 3), EML4 (n = 2), and PLEKHA7 (n = 1), and also three novel fusion partners: CLIP1 (n = 1), KIF5B (n = 1), and KIAA1217 (n = 1). ALK-RCC represents a genetically distinct entity showing a heterogeneous histomorphology, expanded herein to include unreported metanephric adenoma-like and MTSC RCC-like variants. We advocate a routine ALK IHC screening for "unclassifiable RCCs" with heterogeneous features.
Collapse
Affiliation(s)
- Naoto Kuroda
- Department of Diagnostic Pathology, Kochi Red Cross Hospital, Kochi, Japan
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Yuan Gao
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Maria Tretiakova
- Department of Pathology, University of Washington, School of Medicine, Seattle, WA, USA
| | - Yajuan J Liu
- Department of Pathology, University of Washington, School of Medicine, Seattle, WA, USA
| | - Monika Ulamec
- Ljudevit Jurak Pathology Department, University Clinical Hospital Center Sestre Milosrdice, Zagreb; Pathology Department, Medical Faculty, Zagreb, Croatia
| | - Kengo Takeuchi
- Division of Pathology and Pathology Project of Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital of Erlangen, Erlangen, Germany
| | - Christopher Przybycin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute and Glickman Urological Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Soichiro Fushimi
- Department of Pathology, Himeji Red Cross Hospital, Hyogo, Japan
| | - Fumiyoshi Kojima
- Department of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Malthide Sibony
- Anatomie et Cytologie Pathologiques, Cochin Hospital, Paris University, Paris, France
| | - Jen-Fan Hang
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Chen Pan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Asli Yilmaz
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Farshid Siadat
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Emiko Sugawara
- Department of Pathology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Pierre-Alexandre Just
- Anatomie et Cytologie Pathologiques, Cochin Hospital, Paris University, Paris, France
| | - Nikola Ptakova
- Second Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| |
Collapse
|
24
|
Tretiakova MS. Renal Cell Tumors: Molecular Findings Reshaping Clinico-pathological Practice. Arch Med Res 2020; 51:799-816. [PMID: 32839003 DOI: 10.1016/j.arcmed.2020.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
Over the past 20 years, the number of subtypes of renal epithelial cell neoplasia has grown. This growth has resulted from detailed histological and immunohistochemical characterization of these tumors and their correlation with clinical outcomes. Distinctive molecular phenotypes have validated the unique nature of many of these tumors. This growth of unique renal neoplasms has continued after the 2016 World Health Organization (WHO) Classification of Tumours. A consequence is that both the pathologists who diagnose the tumors and the clinicians who care for these patients are confronted with a bewildering array of renal cell carcinoma variants. Many of these variants have important clinical features, i.e. familial or syndromic associations, genomics alterations that can be targeted with systemic therapy, and benignancy of tumors previously classified as carcinomas. Our goal in the review is to provide a practical guide to help recognize these variants, based on small and distinct sets of histological features and limited numbers of immunohistochemical stains, supplemented, as necessary, with molecular features.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
25
|
Ding S, Li S, Zhang S, Li Y. Genetic Alterations and Checkpoint Expression: Mechanisms and Models for Drug Discovery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:227-250. [PMID: 32185713 DOI: 10.1007/978-981-15-3266-5_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this chapter, we will sketch a story that begins with the breakdown of chromosome homeostasis and genomic stability. Genomic alterations may render tumor cells eternal life at the expense of immunogenicity. Although antitumor immunity can be primed through neoantigens or inflammatory signals, tumor cells have evolved countermeasures to evade immune surveillance and strike back by modulating immune checkpoint related pathways. At present, monoclonal antibody drugs targeting checkpoints like PD-1 and CTLA-4 have significantly prolonged the survival of a variety of cancer patients, and thus have marked a great achievement in the history of antitumor therapy. Nevertheless, this is not the end of the story. As the relationship between genomic alteration and checkpoint expression is being delineated though the advances of preclinical animal models and emerging technologies, novel checkpoint targets are on the way to be discovered.
Collapse
Affiliation(s)
- Shuai Ding
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Siqi Li
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Shujie Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China
| | - Yan Li
- The State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Model Animal Research Center of Nanjing University, Nanjing, Jiangsu, 210061, China.
| |
Collapse
|
26
|
Han L, Liu YJ, Ricciotti RW, Mantilla JG. A novel MBTD1-PHF1 gene fusion in endometrial stromal sarcoma: A case report and literature review. Genes Chromosomes Cancer 2020; 59:428-432. [PMID: 32237188 DOI: 10.1002/gcc.22845] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/23/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
The classification of endometrial stromal sarcoma (ESS) has been refined and aided by the discovery of various recurrent gene translocations. Low-grade ESS (LG-ESS) is most commonly characterized by JAZF1-SUZ12 fusions followed by rearrangements involving PHD finger protein-1 (PHF1) and multiple fusion partners, including JAZF1, EPC1, EPC2, and MEAF6. In the present study, integrating anchored polymerase chain reaction and paired-end next-generation ribonucleic acid sequencing, we identified the presence of a novel malignant brain tumor domain-containing 1 (MBTD1)-PHF1 gene fusion in a case of LG-ESS. MBTD1 belongs to the Polycomb gene group, and its fusion with PHF1 is predicted to mediate tumorigenesis through aberrant transcriptional repression. Histology and immunohistochemical studies demonstrated conventional morphology for LG-ESS and clinical follow-up showed no progression of disease after 6 months. These findings help expand the current knowledge on the spectrum of gene rearrangements in the diagnosis of ESS.
Collapse
Affiliation(s)
- Lisa Han
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Yajuan J Liu
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Robert W Ricciotti
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Jose G Mantilla
- Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|