51
|
Linos K, Kerr DA, Baker M, Wong S, Henderson E, Sumegi J, Bridge JA. Superficial malignant ossifying fibromyxoid tumors harboring the rare and recently described ZC3H7B-BCOR and PHF1-TFE3 fusions. J Cutan Pathol 2020; 47:934-945. [PMID: 32352579 DOI: 10.1111/cup.13728] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/16/2020] [Accepted: 04/24/2020] [Indexed: 12/28/2022]
Abstract
Ossifying fibromyxoid tumor (OFMT) is a rare soft tissue neoplasm of uncertain differentiation and intermediate biologic potential. Up to 85% of OFMTs, including benign, atypical, and malignant forms, harbor fusion genes. Most commonly, the PHF1 gene localized to 6p21 is fused with EP400, but other fusion partners, such as MEAF6, EPC1, and JAZF1 have also been described. Herein, we present two rare cases of superficial OFMTs with ZC3H7B-BCOR and the very recently described PHF1-TFE3 fusions. The latter also exhibited moderate to strong diffuse immunoreactivity for TFE3. Reciprocally, this finding expands the entities with TFE3 rearrangements. Accumulation of additional data is necessary to determine if OFMTs harboring these rare fusions feature any reproducible clinicopathologic findings or carry prognostic and/or predictive implications.
Collapse
Affiliation(s)
- Konstantinos Linos
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH and Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Darcy A Kerr
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH and Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Michael Baker
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH and Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Sandra Wong
- Department of Surgery, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Eric Henderson
- Department of Surgery, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Janos Sumegi
- Division of Molecular Pathology, The Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Julia A Bridge
- Division of Molecular Pathology, The Translational Genomics Research Institute, Phoenix, Arizona, USA.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
52
|
Song Y, Yin X, Xia Q, Zheng L, Yao J, Zeng H, Nie L, Gong J, Zhou Q, Chen N. Xp11 translocation renal cell carcinoma with morphological features mimicking multilocular cystic renal neoplasm of low malignant potential: a series of six cases with molecular analysis. J Clin Pathol 2020; 74:171-176. [PMID: 32699116 DOI: 10.1136/jclinpath-2020-206681] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/12/2020] [Accepted: 06/14/2020] [Indexed: 02/05/2023]
Abstract
AIMS Xp11 translocation renal cell carcinoma (RCC) is a distinctive subtype of RCC with TFE3 (Transcription Factor Binding to IGHM Enhancer 3) gene rearrangement. The gross features in most Xp11 translocation RCCs closely resemble clear cell RCCs. In this study, we report six cases of Xp11 translocation RCCs with a unique multicystic architecture, reminiscent of multilocular cystic renal cell neoplasm of low malignant potential (MCRN-LMP). METHODS AND RESULTS Microscopically, the renal mass was well circumscribed with multilocular cystic architecture. The cyst walls and septa were mostly lined by a single layer of cells with clear cytoplasm and low-grade nuclei, reminiscent of MCRN-LMP. Psammoma bodies were detected in four cases. One particular patient was misdiagnosed with benign cysts in local hospitals and led to second operation. Tumour cells were settled according to the track of the first surgical procedure. TFE3 fluorescence in situ hybridization (FISH) assay confirmed the diagnosis of Xp11 translocation RCCs. FISH and RNA sequencing analyses confirmed MED15-TFE3 gene fusion in all six cases. Respective patients were alive, without any recent evidence of disease recurrence and/or metastasis. CONCLUSIONS Here, we introduce a relatively inertia-variant of Xp11 translocation RCC which mimics MCRN-LMP. The distinctive morphological condition is linked to MED15-TFE3 gene fusion. In fact, renal neoplasms with morphological features of MCRN-LMP, especially those containing psammoma bodies, should be routinely evaluated for evidence of TFE3 gene rearrangements.
Collapse
Affiliation(s)
- Yankun Song
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxue Yin
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuyuan Xia
- Pathology, Nanjing Jinling Hospital, Nanjing, China
| | - Linmao Zheng
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Yao
- Radiology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ling Nie
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Gong
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiao Zhou
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ni Chen
- Pathology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
53
|
Williamson SR, Gill AJ, Argani P, Chen YB, Egevad L, Kristiansen G, Grignon DJ, Hes O. Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers: III: Molecular Pathology of Kidney Cancer. Am J Surg Pathol 2020; 44:e47-e65. [PMID: 32251007 PMCID: PMC7289677 DOI: 10.1097/pas.0000000000001476] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) subtypes are increasingly being discerned via their molecular underpinnings. Frequently this can be correlated to histologic and immunohistochemical surrogates, such that only simple targeted molecular assays, or none at all, are needed for diagnostic confirmation. In clear cell RCC, VHL mutation and 3p loss are well known; however, other genes with emerging important roles include SETD2, BAP1, and PBRM1, among others. Papillary RCC type 2 is now known to include likely several different molecular entities, such as fumarate hydratase (FH) deficient RCC. In MIT family translocation RCC, an increasing number of gene fusions are now described. Some TFE3 fusion partners, such as NONO, GRIPAP1, RBMX, and RBM10 may show a deceptive fluorescence in situ hybridization result due to the proximity of the genes on the same chromosome. FH and succinate dehydrogenase deficient RCC have implications for patient counseling due to heritable syndromes and the aggressiveness of FH-deficient RCC. Immunohistochemistry is increasingly available and helpful for recognizing both. Emerging tumor types with strong evidence for distinct diagnostic entities include eosinophilic solid and cystic RCC and TFEB/VEGFA/6p21 amplified RCC. Other emerging entities that are less clearly understood include TCEB1 mutated RCC, RCC with ALK rearrangement, renal neoplasms with mutations of TSC2 or MTOR, and RCC with fibromuscular stroma. In metastatic RCC, the role of molecular studies is not entirely defined at present, although there may be an increasing role for genomic analysis related to specific therapy pathways, such as for tyrosine kinase or MTOR inhibitors.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mutation
- Neoplasm Metastasis
- Neoplastic Syndromes, Hereditary/diagnosis
- Neoplastic Syndromes, Hereditary/genetics
- Neoplastic Syndromes, Hereditary/metabolism
- Neoplastic Syndromes, Hereditary/pathology
- Pathology, Clinical
- Pathology, Molecular
- Prognosis
- Societies, Medical
- Urology
Collapse
Affiliation(s)
- Sean R Williamson
- Department of Pathology and Laboratory Medicine and Henry Ford Cancer Institute, Henry Ford Health System
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - Anthony J Gill
- NSW Health Pathology, Department of Anatomical Pathology
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - David J Grignon
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN
| | - Ondrej Hes
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Pilsen, Czechia
| |
Collapse
|
54
|
An Algorithmic Immunohistochemical Approach to Define Tumor Type and Assign Site of Origin. Adv Anat Pathol 2020; 27:114-163. [PMID: 32205473 DOI: 10.1097/pap.0000000000000256] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunohistochemistry represents an indispensable complement to an epidemiology and morphology-driven approach to tumor diagnosis and site of origin assignment. This review reflects the state of my current practice, based on 15-years' experience in Pathology and a deep-dive into the literature, always striving to be better equipped to answer the age old questions, "What is it, and where is it from?" The tables and figures in this manuscript are the ones I "pull up on the computer" when I am teaching at the microscope and turn to myself when I am (frequently) stuck. This field is so exciting because I firmly believe that, through the application of next-generation immunohistochemistry, we can provide better answers than ever before. Specific topics covered in this review include (1) broad tumor classification and associated screening markers; (2) the role of cancer epidemiology in determining pretest probability; (3) broad-spectrum epithelial markers; (4) noncanonical expression of broad tumor class screening markers; (5) a morphologic pattern-based approach to poorly to undifferentiated malignant neoplasms; (6) a morphologic and immunohistochemical approach to define 4 main carcinoma types; (7) CK7/CK20 coordinate expression; (8) added value of semiquantitative immunohistochemical stain assessment; algorithmic immunohistochemical approaches to (9) "garden variety" adenocarcinomas presenting in the liver, (10) large polygonal cell adenocarcinomas, (11) the distinction of primary surface ovarian epithelial tumors with mucinous features from metastasis, (12) tumors presenting at alternative anatomic sites, (13) squamous cell carcinoma versus urothelial carcinoma, and neuroendocrine neoplasms, including (14) the distinction of pheochromocytoma/paraganglioma from well-differentiated neuroendocrine tumor, site of origin assignment in (15) well-differentiated neuroendocrine tumor and (16) poorly differentiated neuroendocrine carcinoma, and (17) the distinction of well-differentiated neuroendocrine tumor G3 from poorly differentiated neuroendocrine carcinoma; it concludes with (18) a discussion of diagnostic considerations in the broad-spectrum keratin/CD45/S-100-"triple-negative" neoplasm.
Collapse
|
55
|
Ooms AH, Vujanić GM, D’Hooghe E, Collini P, L’Herminé-Coulomb A, Vokuhl C, Graf N, van den Heuvel-Eibrink MM, de Krijger RR. Renal Tumors of Childhood-A Histopathologic Pattern-Based Diagnostic Approach. Cancers (Basel) 2020; 12:cancers12030729. [PMID: 32204536 PMCID: PMC7140051 DOI: 10.3390/cancers12030729] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 03/04/2020] [Accepted: 03/07/2020] [Indexed: 12/15/2022] Open
Abstract
Renal tumors comprise approximately 7% of all malignant pediatric tumors. This is a highly heterogeneous group of tumors, each with its own therapeutic management, outcome, and association with germline predispositions. Histopathology is the key in establishing the correct diagnosis, and therefore pathologists with expertise in pediatric oncology are needed for dealing with these rare tumors. While each tumor shows different histologic features, they do have considerable overlap in cell type and histologic pattern, making the diagnosis difficult to establish, if based on routine histology alone. To this end, ancillary techniques, such as immunohistochemistry and molecular analysis, can be of great importance for the correct diagnosis, resulting in appropriate treatment. To use ancillary techniques cost-effectively, we propose a pattern-based approach and provide recommendations to aid in deciding which panel of antibodies, supplemented by molecular characterization of a subset of genes, are required.
Collapse
Affiliation(s)
- Ariadne H.A.G. Ooms
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Pathan B.V., 3045 PM Rotterdam, The Netherlands
| | | | - Ellen D’Hooghe
- Department of Pathology, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway;
| | - Paola Collini
- Department of Diagnostic Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
| | - Aurore L’Herminé-Coulomb
- Sorbonne Université, Department of Pathology, Hôpital Armand Trousseau, Hopitaux Universitaires Est Parisien, 75012 Paris, France;
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Norbert Graf
- Department of Pediatric Oncology & Hematology, Saarland University, D-66421 Homburg, Germany;
| | | | - Ronald R. de Krijger
- Princess Máxima Center for pediatric oncology, 3584 CS Utrecht, The Netherlands (M.M.v.d.H.-E.)
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
- Correspondence: ; Tel.: +31-088-9727272
| |
Collapse
|
56
|
Herrscher H, Boilève A, Lindner V, Barthélémy P, Hutt É, Pierard L, Kurtz JE, Rioux-Leclercq N, Lang H, Malouf GG. [MiT family translocation renal cell carcinomas: Natural history, molecular features and multidisciplinary management]. Bull Cancer 2020; 107:272-280. [PMID: 32044098 DOI: 10.1016/j.bulcan.2019.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/10/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
MiT family translocation renal cell carcinomas (tRCC) represent a rare subtype of renal cell carcinomas. These tumors have been introduced for the first time in the World Health Classification (WHO) classification of kidney cancers in 2004. tRCC are characterized by reccurent translocations involving members of the MiT family transcription factors, mainly TFE3 and TFEB. The estimated incidence of these tumors is ∼1-5 % among all renal cell carcinomas, with female prodominance. tRCC were initially described in children, and the spectrum has been expanded over time to encompass adolescents and adults. TFE3- and TFEB-rearranged RCC harbor characteristic clinicopathological and immunohistochemical features and fluorescent hybridization in situ is considered the gold standard for their diagnosis, although it has some limitations especially when the partners are located in the vicinity of TFE3. Nephron-sparing surgery is an efficient treatment of localized cases when achievable. In metastatic setting, targeted agents and immunotherapy showed modest efficacy, with response rates and median overall survival inferior to those observed in clear-cell renal cell carcinomas. Management of tRCC necessite a multidisciplinary team and accrual in clinical trials have to be encouraged when possible. Novel biological insights are urgently awaited to better understand the mechanisms associated with kidney oncogenesis in this setting, and ultimately help to identify therapeutic targets.
Collapse
Affiliation(s)
- Hugo Herrscher
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Alice Boilève
- Institut Gustave-Roussy, département de médecine, 67200 Villejuif, France
| | - Véronique Lindner
- Hôpitaux universitaires de Strasbourg, département de pathologie, 67200 Strasbourg, France
| | - Philippe Barthélémy
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Émilie Hutt
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Laure Pierard
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Jean-Emmanuel Kurtz
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France
| | - Nathalie Rioux-Leclercq
- Université de Rennes, service d'anatomie et cytologie pathologiques, Centre Hospitalier Universitaire de Rennes, IRSET, 67200 Rennes, France
| | - Hervé Lang
- Hôpitaux universitaires de Strasbourg, service d'urologie, 35033 Strasbourg, France
| | - Gabriel G Malouf
- Hôpitaux universitaires de Strasbourg, service d'oncologie médicale, 67200 Strasbourg, France.
| |
Collapse
|
57
|
Tretiakova MS, Wang W, Wu Y, Tykodi SS, True L, Liu YJ. Gene fusion analysis in renal cell carcinoma by FusionPlex RNA-sequencing and correlations of molecular findings with clinicopathological features. Genes Chromosomes Cancer 2020; 59:40-49. [PMID: 31400230 DOI: 10.1002/gcc.22798] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/24/2023] Open
Abstract
Translocation renal cell carcinoma (tRCC) affects younger patients and often presents as advanced disease. Accurate diagnosis is required to guide clinical management. Here we evaluate the RNA-sequencing FusionPlex platform with a 115-gene panel including TFE3 and TFEB for tRCC diagnosis and correlate molecular findings with clinicopathological features. We reviewed 996 consecutive RCC cases from our institution over the preceding 7 years and retrieved 17 cases with histological and immunohistochemical features highly suggestive of either TFE3 (n = 16) or TFEB (n = 1). Moderate to strong labeling for TFE3 was present in 15 cases; two cases with weak TFE3 expression were melan-A or cathepsin-K positive. RNA-sequencing detected gene rearrangements in eight cases: PRCC-TFE3 (3), ASPSCR1-TFE3 (2), LUC7L3-TFE3 (1), SFPQ-TFE3 (1), and a novel SETD1B-TFE3 (1). FISH assays of 11 tumors verified six positive cases concordant with FusionPlex analysis results. Two other cases were confirmed by RT-PCR. FusionPlex was superior to FISH by providing precise breakpoints for tRCC-related genes in a single assay and allowing identification of both known and novel fusion partners, thereby facilitating clinicopathological correlations as fusion partners can influence tumor appearance, immunophenotype, and behavior. Cases with partner genes PRCC and novel partner SETD1B were associated with prominent papillary architecture while cases with partner genes ASPSCR1 and LUC7L3 were associated with a predominantly nested/alveolar pattern. The case with SFPQ-TFE3 fusion was characterized by biphasic morphology mimicking TFEB-like translocation RCC. We recommend FusionPlex analysis of RCC in patients under age 50 or when the histologic appearance suggests tRCC.
Collapse
Affiliation(s)
- Maria S Tretiakova
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wenjing Wang
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Yu Wu
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Scott S Tykodi
- Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, Washington
| | - Lawrence True
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington.,Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yajuan J Liu
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Laboratory of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
58
|
Alaghehbandan R, Perez Montiel D, Luis AS, Hes O. Molecular Genetics of Renal Cell Tumors: A Practical Diagnostic Approach. Cancers (Basel) 2019; 12:E85. [PMID: 31905821 PMCID: PMC7017183 DOI: 10.3390/cancers12010085] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022] Open
Abstract
Renal epithelial cell tumors are composed of a heterogeneous group of tumors with variable morphologic, immunohistochemical, and molecular features. A "histo-molecular" approach is now an integral part of defining renal tumors, aiming to be clinically and therapeutically pertinent. Most renal epithelial tumors including the new and emerging entities have distinct molecular and genetic features which can be detected using various methods. Most renal epithelial tumors can be diagnosed easily based on pure histologic findings with or without immunohistochemical examination. Furthermore, molecular-genetic testing can be utilized to assist in arriving at an accurate diagnosis. In this review, we presented the most current knowledge concerning molecular-genetic aspects of renal epithelial neoplasms, which potentially can be used in daily diagnostic practice.
Collapse
Affiliation(s)
- Reza Alaghehbandan
- Department of Pathology, Faculty of Medicine, University of British Columbia, Royal Columbian Hospital, Vancouver, BC V3E 0G9, Canada;
| | - Delia Perez Montiel
- Department of Pathology, Institute Nacional de Cancerologia, INCAN, Mexico DF 14080, Mexico;
| | - Ana Silvia Luis
- Department of Pathology, Centro Hospitalar de Vila Nova de Gaia-Espinho, Vila Nova de Gaia, Cancer Biology and Epigenetics Group (CBEG), IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal;
- Department of Microscopy, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), 4200-072 Porto, Portugal
| | - Ondrej Hes
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzen, 304 60 Pilsen, Czech Republic
| |
Collapse
|
59
|
Whaley RD, Thompson LDR. Primary Thyroid Gland Alveolar Soft Part Sarcoma. Head Neck Pathol 2019; 14:701-706. [PMID: 31782115 PMCID: PMC7413940 DOI: 10.1007/s12105-019-01099-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022]
Abstract
Alveolar soft part sarcoma (ASPS) is a rare soft tissue tumor of unknown histogenesis generally characterized by the der(17)t(X;17)(p11.2;q25) translocation which results in the ASPSCR1-TFE3 gene fusion. Primary ASPS of the thyroid gland has not yet been reported. During oncology follow-up for breast cancer, a pulmonary nodule and thyroid gland mass were identified in a 71-year-old Korean male. Thyroid ultrasound showed a 5.7 cm left thyroid gland mass. After several fine needle aspirations, a thyroid gland lobectomy was performed after documenting only non-caseating granulomatous inflammation in a biopsy of the lung nodule. A 7.6 cm bulging nodular thyroid gland mass was identified, showing significant destructive invasion. Alveolar nests of large polygonal, eosinophilic, granular neoplastic cells were separated by vascularized stroma. Colloid was absent. Tumor necrosis and increased mitoses were identified. The neoplastic cells were positive with TFE3 and CD68, but negative with pancytokeratin, thyroglobulin, TTF-1, napsin-A, calcitonin, PAX8, CAIX, S100 protein, HMB45, SMA, and desmin. FISH confirmed a TFE3 gene rearrangement. The differential includes several primary thyroid gland epithelial neoplasms, paraganglioma, PEComa, melanoma, crystal storage disease, and metastatic carcinomas, especially Xp11 translocation renal cell carcinoma. The patient has refused additional therapy, but is alive without tumor identified (primary or metastatic).
Collapse
Affiliation(s)
- Rumeal D. Whaley
- grid.257413.60000 0001 2287 3919Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN USA
| | - Lester D. R. Thompson
- grid.280062.e0000 0000 9957 7758Department of Pathology, Southern California Permanente Medical Group, 5601 De Soto Avenue, Woodland Hills, CA 91365 USA
| |
Collapse
|
60
|
Xp11 translocation renal cell carcinoma and clear cell renal cell carcinoma with TFE3 strong positive immunostaining: morphology, immunohistochemistry, and FISH analysis. Mod Pathol 2019; 32:1521-1535. [PMID: 31175325 DOI: 10.1038/s41379-019-0283-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 12/18/2022]
Abstract
TFE3 is accepted as a good marker for the diagnosis of Xp11 translocation renal cell carcinoma. However, the significance of TFE3 in other types of renal cell carcinomas remains unclear. We examined the expression of TFE3 using immunohistochemistry by automated Ventana BenchMark XT system in 1818 consecutive renal cell carcinomas and verified the strong positive cases with TFE3 break-apart fluorescence in situ hybridization and RNA sequencing. Among the 27 renal cell carcinomas with TFE3 strong positive immunostaining, 20 cases were diagnosed as Xp11 translocation renal cell carcinoma, and seven cases were diagnosed as clear cell renal cell carcinoma. We further analyzed the morphology, clinicopathological features, and immunohistochemistry markers (CK7, CD117, CD10, P504s, vimentin, CA-IX, AE1/AE3, EMA, HMB45, Melan-A, and cathepsin K) of them. Pale to eosinophilic flocculent cytoplasm and psammomatous calcification were seen only in Xp11 translocation renal cell carcinomas (P < 0.05). Tumor necrosis occurred in all four cases of Xp11 translocation renal cell carcinomas with pT3a stage, which had local recurrence and distant metastasis (two of them died) within 3 years. The expressions of Vimentin, CA-IX, AE1/AE3, and EMA were significantly different between them (P < 0.05). CA-IX was diffusely strong positive in clear cell renal cell carcinomas but negative or focally mild positive in Xp11 translocation renal cell carcinomas. Our study first demonstrates that a very small minority (0.4%) of clear cell renal cell carcinomas with TFE3 strong positive immunostaining, which points out a potential pitfall in diagnosis of Xp11 translocation renal cell carcinomas by TFE3 immunohistochemistry. CA-IX is a good marker to distinguish clear cell renal cell carcinoma with TFE3 strong positive immunostaining from Xp11 translocation renal cell carcinoma. Tumor necrosis could be a potential factor relevant to pT3a stage, which may be a high-risk factor for the patients with Xp11 translocation renal cell carcinomas.
Collapse
|
61
|
Kauffman EC, Lang M, Rais-Bahrami S, Gupta GN, Wei D, Yang Y, Sourbier C, Srinivasan R. Preclinical efficacy of dual mTORC1/2 inhibitor AZD8055 in renal cell carcinoma harboring a TFE3 gene fusion. BMC Cancer 2019; 19:917. [PMID: 31519159 PMCID: PMC6743205 DOI: 10.1186/s12885-019-6096-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022] Open
Abstract
Background Renal cell carcinomas (RCC) harboring a TFE3 gene fusion (TfRCC) represent an aggressive subset of kidney tumors. Key signaling pathways of TfRCC are unknown and preclinical in vivo data are lacking. We investigated Akt/mTOR pathway activation and the preclinical efficacy of dual mTORC1/2 versus selective mTORC1 inhibition in TfRCC. Methods Levels of phosphorylated Akt/mTOR pathway proteins were compared by immunoblot in TfRCC and clear cell RCC (ccRCC) cell lines. Effects of the mTORC1 inhibitor, sirolimus, and the dual mTORC1/2 inhibitor, AZD8055, on Akt/mTOR activation, cell cycle progression, cell viability and cytotoxicity were compared in TfRCC cells. TfRCC xenograft tumor growth in mice was evaluated after 3-week treatment with oral AZD8055, intraperitoneal sirolimus and respective vehicle controls. Results The Akt/mTOR pathway was activated to a similar or greater degree in TfRCC than ccRCC cell lines and persisted partly during growth factor starvation, suggesting constitutive activation. Dual mTORC1/2 inhibition with AZD8055 potently inhibited TfRCC viability (IC50 = 20-50 nM) due at least in part to cell cycle arrest, while benign renal epithelial cells were relatively resistant (IC50 = 400 nM). Maximal viability reduction was greater with AZD8055 than sirolimus (80–90% versus 30–50%), as was the extent of Akt/mTOR pathway inhibition, based on significantly greater suppression of P-Akt (Ser473), P-4EBP1, P-mTOR and HIF1α. In mouse xenograft models, AZD8055 achieved significantly better tumor growth inhibition and prolonged mouse survival compared to sirolimus or vehicle controls. Conclusions Akt/mTOR activation is common in TfRCC and a promising therapeutic target. Dual mTORC1/2 inhibition suppresses Akt/mTOR signaling more effectively than selective mTORC1 inhibition and demonstrates in vivo preclinical efficacy against TFE3-fusion renal cell carcinoma. Electronic supplementary material The online version of this article (10.1186/s12885-019-6096-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eric C Kauffman
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Departments of Urology and Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Soroush Rais-Bahrami
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Department of Urology and Department of Radiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35294, USA
| | - Gopal N Gupta
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Department of Urology, Loyola University Medical Center, Chicago, IL, 60153, USA
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Youfeng Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.
| |
Collapse
|
62
|
Gandhi JS, Malik F, Amin MB, Argani P, Bahrami A. MiT family translocation renal cell carcinomas: A 15th anniversary update. Histol Histopathol 2019; 35:125-136. [PMID: 31489603 DOI: 10.14670/hh-18-159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microphthalmia (MiT) family translocation renal cell carcinomas (RCCs) are a heterogeneous category of renal tumors which all express MiT transcription factors, typically from chromosomal translocation and rarely from gene amplification. This tumor family has two major subtypes [i.e., Xp11 translocation RCC and t(6;11) RCC] and several related neoplasms (i.e., TFEB amplification RCC and melanotic Xp11 translocation renal cancers). Increased understanding of the clinical, pathological, molecular and prognostic heterogeneity of these tumors, since their official recognition in 2004, provides the opportunity to identify prognostic biomarkers and to understand the reasons for tumor aggression. We will review the literature from the past 15 years and highlight the need for a greater understanding of the molecular mechanisms underpinning heterogeneous tumor behavior.
Collapse
Affiliation(s)
- Jatin S Gandhi
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Faizan Malik
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mahul B Amin
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Pedram Argani
- Department of Pathology, John Hopkins University, Baltimore, MD, USA
| | - Armita Bahrami
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, USA.,Departments of Pathology and Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
63
|
Caliò A, Segala D, Munari E, Brunelli M, Martignoni G. MiT Family Translocation Renal Cell Carcinoma: from the Early Descriptions to the Current Knowledge. Cancers (Basel) 2019; 11:E1110. [PMID: 31382581 PMCID: PMC6721505 DOI: 10.3390/cancers11081110] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/27/2022] Open
Abstract
The new category of MiT family translocation renal cell carcinoma has been included into the World Health Organization (WHO) classification in 2016. The MiT family translocation renal cell carcinoma comprises Xp11 translocation renal cell carcinoma harboring TFE3 gene fusions and t(6;11) renal cell carcinoma harboring TFEB gene fusion. At the beginning, they were recognized in childhood; nevertheless, it has been demonstrated that these neoplasms can occur in adults as well. In the nineties, among Xp11 renal cell carcinoma, ASPL, PRCC, and SFPQ (PSF) were the first genes recognized as partners in TFE3 rearrangement. Recently, many other genes have been identified, and a wide spectrum of morphologies has been described. For this reason, the diagnosis may be challenging based on the histology, and the differential diagnosis includes the most common renal cell neoplasms and pure epithelioid PEComa/epithelioid angiomyolipoma of the kidney. During the last decades, many efforts have been made to identify immunohistochemical markers to reach the right diagnosis. To date, staining for PAX8, cathepsin K, and melanogenesis markers are the most useful identifiers. However, the diagnosis requires the demonstration of the chromosomal rearrangement, and fluorescent in situ hybridization (FISH) is considered the gold standard. The outcome of Xp11 translocation renal cell carcinoma is highly variable, with some patients surviving decades with indolent disease and others dying rapidly of progressive disease. Despite most instances of t(6;11) renal cell carcinoma having an indolent clinical course, a few published cases demonstrate aggressive behavior. Recently, renal cell carcinomas with TFEB amplification have been described in connection with t(6;11) renal cell carcinoma. Those tumors appear to be associated with a more aggressive clinical course. For the aggressive cases of MiT family translocation carcinoma, the optimal therapy remains to be determined; however, new target therapies seem to be promising, and the search for predictive markers is mandatory.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy
| | - Diego Segala
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda 37019, Italy
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Hospital, Negrar 37024, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona 37134, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda 37019, Italy.
| |
Collapse
|
64
|
Yu W, Wang Y, Rao Q, Jiang Y, Zhang W, Li Y. Xp11.2 translocation renal neoplasm with features of TFE3 rearrangement associated renal cell carcinoma and Xp11 translocation renal mesenchymal tumor with melanocytic differentiation harboring NONO-TFE3 fusion gene. Pathol Res Pract 2019; 215:152521. [PMID: 31277953 DOI: 10.1016/j.prp.2019.152521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
Xp11.2 translocation/TFE3 rearrangement-associated renal cell carcinoma (RCC) and Xp11 translocation renal mesenchymal tumor are distinct tumor entity. To broaden the spectrum of Xp11 neoplasms, we investigated a novel tumor exhibiting morphologies overlapping Xp11.2 translocation/TFE3 rearrangement-associated RCC and the mesenchymal counterpart with melanocytic differentiation by immunohistochemistry, fluorescence in situ hybridization (FISH) and RNA sequencing, as well as literature review. Histologically, the tumor was composed of three different types of tumor cells, including a large proportion of clear cells, small round cells, and a few spindle cells, presenting a relatively clear border in the majority area. The nuclei of all tumor cells showed extensively and strong positive expressions of TFE3. Whereas, the clear cells positively expressed the RCC-related markers including PAX8, RCC marker and CD10, and negatively expressed HMB45; On the contrary, the small round cells and spindle cells positively expressed melanocytic marker HMB45, and negatively expressed PAX8, RCC marker and CD10. The ki67 index was higher in the small round cells and spindle cells than that in the clear cells. FISH revealed the rearrangement of TFE3 gene in all the three types of cells. The NONO-TFE3 fusion gene was detected in all tumor cells by RNA sequencing. This unique Xp11 translocation-associated neoplasm might represent a distinct entity overlapping Xp11 translocation RCC and the mesenchymal counterpart with melanocytic differentiation, broadening the spectrum of Xp11 neoplasms. The patient died of tumor recurrence and lung metastasis after seven months after the surgery suggesting those tumors have an unfavorable prognosis.
Collapse
Affiliation(s)
- Wenjuan Yu
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Yuewei Wang
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qiu Rao
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yanxia Jiang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Wei Zhang
- Department of Pathology, 971 Hospital of Navy of Chinese People's Liberation Army, NO. 22 Minjiang Rd. Qingdao, 266071, China.
| | - Yujun Li
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
65
|
Linehan WM, Schmidt LS, Crooks DR, Wei D, Srinivasan R, Lang M, Ricketts CJ. The Metabolic Basis of Kidney Cancer. Cancer Discov 2019; 9:1006-1021. [PMID: 31088840 DOI: 10.1158/2159-8290.cd-18-1354] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/19/2019] [Accepted: 03/22/2019] [Indexed: 01/27/2023]
Abstract
Kidney cancer is not a single disease but represents several distinct types of cancer that have defining histologies and genetic alterations and that follow different clinical courses and have different responses to therapy. Mutation of genes associated with kidney cancer, such as VHL, FLCN, TFE3, FH, or SDHB, dysregulates the tumor's responses to changes in oxygen, iron, nutrient, or energy levels. The identification of these varying genetic bases of kidney cancer has increased our understanding of the biology of this cancer, allowing the development of targeted therapies and the appreciation that it is a cancer driven by metabolic alterations. SIGNIFICANCE: Kidney cancer is a complex disease composed of different types of cancer that present with different histologies, clinical courses, genetic changes, and responses to therapy. This review describes the known genetic changes within kidney cancer, how they alter tumor metabolism, and how these metabolic changes can be therapeutically targeted.
Collapse
Affiliation(s)
- W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.,Basic Science Program, Frederick Laboratory for Cancer Research, Frederick, Maryland
| | - Daniel R Crooks
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
66
|
Xie L, Zhang Y, Wu CL. Microphthalmia family of transcription factors associated renal cell carcinoma. Asian J Urol 2019; 6:312-320. [PMID: 31768316 PMCID: PMC6872788 DOI: 10.1016/j.ajur.2019.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/25/2018] [Accepted: 11/06/2018] [Indexed: 12/11/2022] Open
Abstract
The microphthalmia (MiT) subfamily of transcription factors includes TFE3, TFEB, TFEC, and MITF. In the 2016 World Health Organization classification, MiT family translocation renal cell carcinoma (tRCC) including Xp11 tRCC and t(6;11) RCC, was newly defined as an RCC subtype. Xp11 and t(6;11) RCC are characterized by the rearrangement of the MiT transcription factors TFE3 and TFEB, respectively. Recent studies identified the fusion partner-dependent clinicopathological and immunohistochemical features in TFE3-rearranged RCC. Furthermore, RCC with TFEB amplification, melanotic MiT family translocation neoplasms, was identified may as a unique subtype of MiT family associated renal neoplasms, along with MITF associated RCC. In this review, we will collect available literature of these newly-described RCCs, analyze their clinicopathological and immunohistochemical features, and summarize their molecular and genetic evidences. We expect this review would be beneficial for the understanding of these rare subtypes of RCCs, and eventually promote clinical management strategies.
Collapse
Affiliation(s)
- Ling Xie
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pathology and Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yifen Zhang
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chin-Lee Wu
- Department of Pathology and Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
67
|
NEAT1-TFE3 and KAT6A-TFE3 renal cell carcinomas, new members of MiT family translocation renal cell carcinoma. Mod Pathol 2019; 32:710-716. [PMID: 30622287 PMCID: PMC6486435 DOI: 10.1038/s41379-018-0191-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 12/31/2022]
Abstract
Microphthalmia-associated transcription factor (MiT) family translocation renal cell carcinoma harbors variable gene fusions involving either TFE3 or TFEB genes. Multiple 5' fusion partners for TFE3 have been reported, including ASPSCR1, CLTC, DVL2, LUC7L3, KHSRP, PRCC, PARP14, NONO, SFPQ1, MED15, and RBM10. Each of these fusion genes activates TFE3 transcription which can be detected by immunostaining. Using targeted RNA-sequencing, TFE3 fusion gene partners were identified in 5 cases of TFE3 immunohistochemistry positive translocation renal cell carcinoma. Three cases demonstrated known fusions: ASPSCR1-TFE3, MED15-TFE3 and RBM10-TFE3. However, two cases showed unreported NEAT1-TFE3 and KAT6A-TFE3 fusion transcripts. The NEAT1-TFE3 RCC arose in a 59-year-old male; which demonstrated overlapping morphological features seen in NEAT2(MALAT1)-TFEB t(6;11) renal cell carcinoma, including biphasic alveolar/nested tumor cells with eosinophilic cytoplasm. The KAT6A-TFE3 renal cell carcinoma demonstrated typical morphological features of TFE3/Xp11 renal cell carcinoma including papillae, eosinophilic cytoplasm with focal clearing and abundant psammoma bodies. KAT6A gene fusion was reported in some cases of acute myeloid leukemia, which has not been previously reported in solid tumors. This report highlights the genetic complexity of TFE3 translocation renal cell carcinoma; and RNA-sequencing is a powerful approach for elucidating the underlying genetic alterations.
Collapse
|
68
|
TFE3 fusions escape from controlling of mTOR signaling pathway and accumulate in the nucleus promoting genes expression in Xp11.2 translocation renal cell carcinomas. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:119. [PMID: 30849994 PMCID: PMC6408813 DOI: 10.1186/s13046-019-1101-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
Abstract
Background Xp11.2 translocation renal cell carcinoma (tRCC) is mainly caused by translocation of the TFE3 gene located on chromosome Xp11.2 and is characterized by overexpression of the TFE3 fusion gene. Patients are diagnosed with tRCC usually before 45 years of age with poor prognosis. We investigated this disease using two tRCC cell lines, UOK109 and UOK120, in this study. Methods The purpose of this study was to investigate the pathogenic mechanism of TFE3 fusions in tRCC based on its subcellular localization, nuclear translocation and transcriptional activity. The expression of TFE3 fusions and other related genes were analyzed by quantitative reverse transcription PCR (qRT-PCR) and Western blot. The subcellular localization of TFE3 was determined using immunofluorescence. The transcriptional activity of TFE3 fusions was measured using a luciferase reporter assay and ChIP analysis. In some experiments, TFE3 fusions were depleted by RNAi or gene knockdown. The TFE3 fusion segments were cloned into a plasmid expression system for expression in cells. Results Our results demonstrated that TFE3 fusions were overexpressed in tRCC with a strong nuclear retention irrespective of treatment with an mTORC1 inhibitor or not. TFE3 fusions lost its co-localization with lysosomal proteins and decreased its interaction with the chaperone 14–3-3 proteins in UOK109 and UOK120 cells. However, the fusion segments of TFE3 could not translocate to the nucleus and inhibition of Gsk3β could increase the cytoplasmic retention of TFE3 fusions. Both the luciferase reporter assay and ChIP analysis demonstrated that TFE3 fusions could bind to the promoters of the target genes as a wild-type TFE3 protein. Knockdown of TFE3 results in decreased expression of those genes responsible for lysosomal biogenesis and other target genes. The ChIP-seq data further verified that, in addition to lysosomal genes, TFE3 fusions could regulate genes involved in cellular responses to hypoxic stress and transcription. Conclusions Our results indicated that the overexpressed TFE3 fusions were capable of escaping from the control by the mTOR signaling pathway and were accumulated in the nucleus in UOK109 and UOK120 cells. The nuclear retention of TFE3 fusions promoted the expression of lysosomal genes and other target genes, facilitating cancer cell resistance against an extreme environment. Electronic supplementary material The online version of this article (10.1186/s13046-019-1101-7) contains supplementary material, which is available to authorized users.
Collapse
|
69
|
Perera RM, Di Malta C, Ballabio A. MiT/TFE Family of Transcription Factors, Lysosomes, and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2019; 3:203-222. [PMID: 31650096 PMCID: PMC6812561 DOI: 10.1146/annurev-cancerbio-030518-055835] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells have an increased demand for energy sources to support accelerated rates of growth. When nutrients become limiting, cancer cells may switch to nonconventional energy sources that are mobilized through nutrient scavenging pathways involving autophagy and the lysosome. Thus, several cancers are highly reliant on constitutive activation of these pathways to degrade and recycle cellular materials. Here, we focus on the MiT/TFE family of transcription factors, which control transcriptional programs for autophagy and lysosome biogenesis and have emerged as regulators of energy metabolism in cancer. These new findings complement earlier reports that chromosomal translocations and amplifications involving the MiT/TFE genes contribute to the etiology and pathophysiology of renal cell carcinoma, melanoma, and sarcoma, suggesting pleiotropic roles for these factors in a wider array of cancers. Understanding the interplay between the oncogenic and stress-adaptive roles of MiT/TFE factors could shed light on fundamental mechanisms of cellular homeostasis and point to new strategies for cancer treatment.
Collapse
Affiliation(s)
- Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
70
|
Schneider M, Dinkelborg K, Xiao X, Chan-Smutko G, Hruska K, Huang D, Sagar P, Harisinghani M, Iliopoulos O. Early onset renal cell carcinoma in an adolescent girl with germline FLCN exon 5 deletion. Fam Cancer 2019. [PMID: 28623476 DOI: 10.1007/s10689-017-0008-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Birt-Hogg-Dube (BHD) disease is an autosomal dominant cancer syndrome characterized by benign skin tumors, renal cancer and spontaneous pneumothorax and is caused by mutations in the Folliculin (FLCN) gene. Benign skin tumors and pneumothorax occur in the majority of patients affected by BHD syndrome, but only 30-45% of them develop renal cell carcinoma (RCC) with a median age of diagnosis at 48. The earliest onset of RCC in a BHD patient has been reported at age 20. Here we report a case of a 14 year-old patient with germline FLCN mutation leading to an early-onset bulky RCC that could not be classified strictly according to existing histological types. Germline genetic testing revealed a deletion at FLCN exon 5. The father of the patient was identified as the asymptomatic carrier. We report the youngest patient with BHD-related RCC. This early onset presentation supports genetic testing of at-risk patients and initiation of imaging surveillance for RCC in early adolescence. In addition, future studies are necessary to understand the determinants of reduced penetrance in BHD disease.
Collapse
Affiliation(s)
- Meike Schneider
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, 02115, USA.,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA
| | - Katja Dinkelborg
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA.,Harvard Medical School, Boston, MA, 02115, USA.,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA
| | - Xiuli Xiao
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA.,Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Gayun Chan-Smutko
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | | | | | - Pallavi Sagar
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mukesh Harisinghani
- Department of Radiology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Othon Iliopoulos
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA. .,Harvard Medical School, Boston, MA, 02115, USA. .,MGH Center for Cancer Research, 149 13th Street, Charlestown, MA, 02129, USA.
| |
Collapse
|
71
|
Damayanti NP, Budka JA, Khella HWZ, Ferris MW, Ku SY, Kauffman E, Wood AC, Ahmed K, Chintala VN, Adelaiye-Ogala R, Elbanna M, Orillion A, Chintala S, Kao C, Linehan WM, Yousef GM, Hollenhorst PC, Pili R. Therapeutic Targeting of TFE3/IRS-1/PI3K/mTOR Axis in Translocation Renal Cell Carcinoma. Clin Cancer Res 2018; 24:5977-5989. [PMID: 30061365 PMCID: PMC6279468 DOI: 10.1158/1078-0432.ccr-18-0269] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 01/05/2023]
Abstract
PURPOSE Translocation renal cell carcinoma (tRCC) represents a rare subtype of kidney cancer associated with various TFE3, TFEB, or MITF gene fusions that are not responsive to standard treatments for RCC. Therefore, the identification of new therapeutic targets represents an unmet need for this disease. EXPERIMENTAL DESIGN We have established and characterized a tRCC patient-derived xenograft, RP-R07, as a novel preclinical model for drug development by using next-generation sequencing and bioinformatics analysis. We then assessed the therapeutic potential of inhibiting the identified pathway using in vitro and in vivo models. RESULTS The presence of a SFPQ-TFE3 fusion [t(X;1) (p11.2; p34)] with chromosomal break-points was identified by RNA-seq and validated by RT-PCR. TFE3 chromatin immunoprecipitation followed by deep sequencing analysis indicated a strong enrichment for the PI3K/AKT/mTOR pathway. Consistently, miRNA microarray analysis also identified PI3K/AKT/mTOR as a highly enriched pathway in RP-R07. Upregulation of PI3/AKT/mTOR pathway in additional TFE3-tRCC models was confirmed by significantly higher expression of phospho-S6 (P < 0.0001) and phospho-4EBP1 (P < 0.0001) in established tRCC cell lines compared with clear cell RCC cells. Simultaneous vertical targeting of both PI3K/AKT and mTOR axis provided a greater antiproliferative effect both in vitro (P < 0.0001) and in vivo (P < 0.01) compared with single-node inhibition. Knockdown of TFE3 in RP-R07 resulted in decreased expression of IRS-1 and inhibited cell proliferation. CONCLUSIONS These results identify TFE3/IRS-1/PI3K/AKT/mTOR as a potential dysregulated pathway in TFE3-tRCC, and suggest a therapeutic potential of vertical inhibition of this axis by using a dual PI3K/mTOR inhibitor for patients with TFE3-tRCC.
Collapse
Affiliation(s)
- Nur P Damayanti
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Justin A Budka
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Heba W Z Khella
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science at the Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Mary W Ferris
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Sheng Yu Ku
- Department of Pharmacology & Therapeutics, Roswell Park Cancer Institute, Buffalo, New York
| | - Eric Kauffman
- Department of Urology and Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, New York
| | - Anthony C Wood
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Khunsha Ahmed
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Venkata Nithinsai Chintala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Remi Adelaiye-Ogala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - May Elbanna
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Ashley Orillion
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Sreenivasulu Chintala
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Chinghai Kao
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | | | - George M Yousef
- Department of Laboratory Medicine and the Keenan Research Centre for Biomedical Science at the Li KaShing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University School of Medicine, Bloomington, Indiana
| | - Roberto Pili
- Genitourinary Program, Division of Hematology & Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana.
| |
Collapse
|
72
|
Xiang H, Zhao N, Chen Z. The detection of PAX8 in human upper urinary tract urothelial carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:5394-5400. [PMID: 31949621 PMCID: PMC6963013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/11/2018] [Indexed: 06/10/2023]
Abstract
Upper urinary tract urothelial carcinomas (UUT-UCs) are defined as malignant neoplasms of the urothelium from the upper urinary tract, including renal calyces, the renal pelvis and the distal ureter. The natural attributes of UUT-UCs differ from those of bladder cancer. The aim of the present study was to investigate PAX8 expression in the normal urothelium and in urothelial carcinomas (UCs) of the upper urinary tract. Immunohistochemistry was conducted in 35 cases of renal pelvic and 30 cases of ureteral papillary UCs and the adjacent normal urothelium respectively. PAX8 mRNA expression was evaluated by RT-PCR in a different set of normal urothelial mucosa of the urinary tract and UUT-UCs. In immunohistochemical studies, the positive rates of PAX8 staining in UCs of the renal pelvis and ureter were 17% and 6.6% respectively, presenting focally positive in most cases, while the positive rates in the adjacent normal epithelia of the pelvis and ureter were 100% and 93% respectively. PAX8 mRNA was detected in all of the tumors and adjacent normal urothelial mucosa specimens of the upper urinary tract. 4 types of PAX8 isoforms, PAX8a, PAX8b, PAX8c and PAX8e, were detected in UUT-UCs in this study. As in bladder cancer, PAX8 expression was highly heterogeneous in terms of the splicing mRNA isoforms, with the different isoforms differentially expressed in the UUT-UCs. Among the 4 types of PAX8 isoforms, the PAX8e isoform was found in almost all UUT-UCs tumor tissues, but the PAX8d isoform was not detected in UUT-UCs that were different from the transcriptional splicing patterns of PAX8 in bladder cancer reported in the literature. In addition, the above 4 types of PAX8 splicing isoforms were simultaneously detected in almost all of the normal mucosal epithelia of the upper urinary tract, which was very different from that of bladder mucosa. Further studies are suggested to reveal whether or not the differences in natural attributes between UCs of the upper and lower urinary tracts are related to their PAX8 transcriptional splicing patterns.
Collapse
Affiliation(s)
- Hua Xiang
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, China
| | - Na Zhao
- Institute of Zoology, The Chinese Academy of SciencesBeijing, China
| | - Zhen Chen
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhou, Zhejiang Province, China
| |
Collapse
|
73
|
Cytogenetic and immunohistochemical study of 42 pigmented microcystic chromophobe renal cell carcinoma (PMChRCC). Virchows Arch 2018; 473:209-217. [DOI: 10.1007/s00428-018-2389-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 05/12/2018] [Accepted: 06/11/2018] [Indexed: 01/25/2023]
|
74
|
Recommendations for the Management of Rare Kidney Cancers. Eur Urol 2017; 72:974-983. [DOI: 10.1016/j.eururo.2017.06.040] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/30/2017] [Indexed: 12/31/2022]
|
75
|
Al-Maghrabi JA, Butt NS, Anfinan N, Sait K, Sait H, Marzouki A, Khabaz MN. Infrequent Immunohistochemical Expression of Napsin A in Endometrial Carcinomas. Appl Immunohistochem Mol Morphol 2017; 25:632-638. [DOI: 10.1097/pai.0000000000000350] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
76
|
Xiao X, Hu R, Deng FM, Shen SS, Yang XJ, Wu CL. Practical Applications of Immunohistochemistry in the Diagnosis of Genitourinary Tumors. Arch Pathol Lab Med 2017; 141:1181-1194. [DOI: 10.5858/arpa.2016-0530-ra] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Context.—Pathologic diagnosis of tumors in the genitourinary system can be challenging based on morphology alone, particularly when diagnostic material is limited, such as in core biopsies. Immunohistochemical stain can be a useful tool to aid in the diagnosis.Objective.—To provide an update on practical applications and interpretation of immunohistochemical stains in the diagnosis of tumors in prostate, kidney, bladder, and testis. We particularly focus on difficult differential diagnoses, providing our insights in frequently encountered challenging situations. Commonly used immunohistochemical panels are discussed.Data Sources.—Review of literature and our own experience.Conclusion.—Immunohistochemical stain is a valuable tool in the diagnosis of genitourinary tumors when appropriately used.
Collapse
|
77
|
Inamura K. Translocation Renal Cell Carcinoma: An Update on Clinicopathological and Molecular Features. Cancers (Basel) 2017; 9:cancers9090111. [PMID: 28850056 PMCID: PMC5615326 DOI: 10.3390/cancers9090111] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/24/2017] [Accepted: 08/24/2017] [Indexed: 12/19/2022] Open
Abstract
Microphthalmia-associated transcription (MiT) family translocation renal cell carcinoma (tRCC) comprises Xp11 tRCC and t(6;11) RCC. Due to the presence of fusion genes, Xp11 tRCC and t(6;11) RCC are also known as TFE3- and TFEB-rearranged RCC, respectively. TFE3 and TFEB belong to the MiT family, which regulates melanocyte and osteoclast differentiation, and TFE3- and TFEB-rearranged RCC show characteristic clinicopathological and immunohistochemical features. Recent studies identified the fusion partner-dependent clinicopathological and immunohistochemical features in TFE3-rearranged RCC. Furthermore, RCC with chromosome 6p amplification, including TFEB, was identified as a unique subtype of RCC, along with ALK-rearranged RCC. This review summarizes these recent advancements in our tRCC-related knowledge.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
78
|
Slade L, Pulinilkunnil T. The MiTF/TFE Family of Transcription Factors: Master Regulators of Organelle Signaling, Metabolism, and Stress Adaptation. Mol Cancer Res 2017; 15:1637-1643. [PMID: 28851811 DOI: 10.1158/1541-7786.mcr-17-0320] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/08/2017] [Accepted: 08/24/2017] [Indexed: 11/16/2022]
Abstract
The microphthalmia family (MITF, TFEB, TFE3, and TFEC) of transcription factors is emerging as global regulators of cancer cell survival and energy metabolism, both through the promotion of lysosomal genes as well as newly characterized targets, such as oxidative metabolism and the oxidative stress response. In addition, MiT/TFE factors can regulate lysosomal signaling, which includes the mTORC1 and Wnt/β-catenin pathways, which are both substantial contributors to oncogenic signaling. This review describes recent discoveries in MiT/TFE research and how they impact multiple cancer subtypes. Furthermore, the literature relating to TFE-fusion proteins in cancers and the potential mechanisms through which these genomic rearrangements promote tumorigenesis is reviewed. Likewise, the emerging function of the Folliculin (FLCN) tumor suppressor in negatively regulating the MiT/TFE family and how loss of this pathway promotes cancer is examined. Recent reports are also presented that relate to the role of MiT/TFE-driven lysosomal biogenesis in sustaining cancer cell metabolism and signaling in nutrient-limiting conditions. Finally, a discussion is provided on the future directions and unanswered questions in the field. In summary, the research surrounding the MiT/TFE family indicates that these transcription factors are promising therapeutic targets and biomarkers for cancers that thrive in stressful niches. Mol Cancer Res; 15(12); 1637-43. ©2017 AACR.
Collapse
Affiliation(s)
- Logan Slade
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, New Brunswick, Canada
| | - Thomas Pulinilkunnil
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Dalhousie University, Dalhousie Medicine New Brunswick, New Brunswick, Canada.
| |
Collapse
|
79
|
Saleeb RM, Srigley JR, Sweet J, Doucet C, Royal V, Chen YB, Brimo F, Evans A. Melanotic MiT family translocation neoplasms: Expanding the clinical and molecular spectrum of this unique entity of tumors. Pathol Res Pract 2017; 213:1412-1418. [PMID: 28969862 DOI: 10.1016/j.prp.2017.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/13/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022]
Abstract
MiT family translocation tumors are a group of neoplasms characterized by translocations involving MiT family transcription factors. The translocation renal cell carcinomas, TFE3 (Xp11.2) and TFEB (t6;11) are known members of this family. Melanotic Xp11 translocation renal cancer is a more recently described entity. To date only 14 cases have been described. It is characterized by a distinct set of features including a nested epithelioid morphology, melanin pigmentation, labeling for markers of melanocytic differentiation, lack of labeling for markers of renal tubular differentiation, predominance in a younger age population and association with aggressive clinical behavior. There are noted similarities between that entity and TFE3 associated PEComas. There are no cases reported of equivalent melanotic TFEB translocation renal cancer. We report 2 rare cases of melanotic translocation renal neoplasms. The first is a melanotic TFE3 translocation renal cancer with an indolent clinical course, occurring in a patient more than 3-decades older than the usual average age in which such tumors have been described. The other case is, to our knowledge, the first reported melanotic TFEB translocation cancer of the kidney. Both cases exhibit the same H&E morphology as previously reported in melanotic translocation renal cancers and label accordingly with HMB45 and Melan-A. While the TFE3 melanotic tumor lacked any evidence of renal tubular differentiation, the TFEB melanotic cancer exhibited some staining for renal tubular markers. Based on the unique features noted above, these two cases expand the clinical and molecular spectrum of the melanotic translocation renal cancers.
Collapse
Affiliation(s)
- Rola M Saleeb
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - John R Srigley
- Department of Pathology, Credit Valley Hospital, Mississauga, Ontario, Canada; Department of Pathology and molecular medicine, McMaster University, Hamilton, Ontario, Canada
| | - Joan Sweet
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Toronto General Hospital, Toronto, Ontario, Canada
| | - Cedric Doucet
- McGill University Health Center, Montreal, Quebec, Canada
| | - Virginie Royal
- Pathology Department, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, United States
| | - Fadi Brimo
- Department of Pathology, McGill University Health Center, Montreal, Quebec, Canada.
| | - Andrew Evans
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Pathology, Toronto General Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
80
|
TFEB-amplified Renal Cell Carcinomas: An Aggressive Molecular Subset Demonstrating Variable Melanocytic Marker Expression and Morphologic Heterogeneity. Am J Surg Pathol 2017; 40:1484-1495. [PMID: 27565001 DOI: 10.1097/pas.0000000000000720] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Renal cell carcinomas (RCCs) with the t(6;11)(p21;q12) chromosome translocation are low-grade RCC which often occur in young patients. They typically feature an unusual biphasic morphology characterized by nests of larger epithelioid cells surrounding intraluminal collections of smaller cells clustered around basement membrane material. The t(6;11)(p21;q12) translocation fuses the Alpha (MALAT1) gene with the TFEB transcription factor gene, resulting in upregulated expression of intact native TFEB that drives the aberrant expression of melanocytic markers which is a hallmark of this distinctive neoplasm. We now report 8 cases of RCC, which demonstrate TFEB gene amplification (6 without TFEB rearrangement, 2 with concurrent TFEB rearrangement) and demonstrate downstream consequences of TFEB overexpression. Like the unamplified t(6;11) RCC, all TFEB-amplified RCC were associated with aberrant melanocytic marker expression. However, several differences between TFEB-amplified RCC and the usual unamplified t(6;11) RCC are evident. First, TFEB-amplified RCC occurred in older patients (median age, 64.5 y) compared with unamplified t(6;11) RCC (median age, 31 y). Second, the morphology of TFEB-amplified RCC is not entirely distinctive, frequently featuring nests of high-grade epithelioid cells with eosinophilic cytoplasm associated with pseudopapillary formation and necrosis, or true papillary formations. These patterns raise the differential diagnosis of high-grade clear cell and papillary RCC. Third, TFEB and melanocytic marker expression was more variable within the TFEB-amplified RCC. TFEB protein expression by immunohistochemistry was detectable in 6 of 8 cases. While all 8 cases expressed melan-A, only 5 of 8 expressed cathepsin K and only 3 of 8 expressed HMB45. Fourth, the TFEB-amplified RCC were associated with a more aggressive clinical course; 3 of 8 cases presented with advanced stage or metastatic disease, 2 subsequently developed metastatic disease, whereas the other 3 cases had minimal/no follow-up. Our results are corroborated by scant data reported on 6 TFEB-amplified RCC in the literature, gleaned from 1 case report, 1 abstract, and 4 individual cases identified within 2 genomic studies of large cohorts of RCC. In summary, TFEB-amplified RCC represent a distinct molecular subtype of high-grade adult RCC associated with aggressive clinical behavior, variable morphology, and aberrant melanocytic marker expression.
Collapse
|
81
|
TFE3-Fusion Variant Analysis Defines Specific Clinicopathologic Associations Among Xp11 Translocation Cancers. Am J Surg Pathol 2017; 40:723-37. [PMID: 26975036 DOI: 10.1097/pas.0000000000000631] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Xp11 translocation cancers include Xp11 translocation renal cell carcinoma (RCC), Xp11 translocation perivascular epithelioid cell tumor (PEComa), and melanotic Xp11 translocation renal cancer. In Xp11 translocation cancers, oncogenic activation of TFE3 is driven by the fusion of TFE3 with a number of different gene partners; however, the impact of individual fusion variant on specific clinicopathologic features of Xp11 translocation cancers has not been well defined. In this study, we analyze 60 Xp11 translocation cancers by fluorescence in situ hybridization using custom bacterial artificial chromosome probes to establish their TFE3 fusion gene partner. In 5 cases RNA sequencing was also used to further characterize the fusion transcripts. The 60 Xp11 translocation cancers included 47 Xp11 translocation RCC, 8 Xp11 translocation PEComas, and 5 melanotic Xp11 translocation renal cancers. A fusion partner was identified in 53/60 (88%) cases, including 18 SFPQ (PSF), 16 PRCC, 12 ASPSCR1 (ASPL), 6 NONO, and 1 DVL2. We provide the first morphologic description of the NONO-TFE3 RCC, which frequently demonstrates subnuclear vacuoles leading to distinctive suprabasal nuclear palisading. Similar subnuclear vacuolization was also characteristic of SFPQ-TFE3 RCC, creating overlapping features with clear cell papillary RCC. We also describe the first RCC with a DVL2-TFE3 gene fusion, in addition to an extrarenal pigmented PEComa with a NONO-TFE3 gene fusion. Furthermore, among neoplasms with the SFPQ-TFE3, NONO-TFE3, DVL2-TFE3, and ASPL-TFE3 gene fusions, the RCCs are almost always PAX8 positive, cathepsin K negative by immunohistochemistry, whereas the mesenchymal counterparts (Xp11 translocation PEComas, melanotic Xp11 translocation renal cancers, and alveolar soft part sarcoma) are PAX8 negative, cathepsin K positive. These findings support the concept that despite an identical gene fusion, the RCCs are distinct from the corresponding mesenchymal neoplasms, perhaps due to the cellular context in which the translocation occurs. We corroborate prior data showing that the PRCC-TFE3 RCCs are the only known Xp11 translocation RCC molecular subtype that are consistently cathepsin K positive. In summary, our data expand further the clinicopathologic features of cancers with specific TFE3 gene fusions and should allow for more meaningful clinicopathologic associations to be drawn.
Collapse
|
82
|
Fang Y, Bao W, Rao Q, Wang X, Xia Q, Shen Q, Zhou X, Yao B. TFE3 regulates renal adenocarcinoma cell proliferation via activation of the mTOR pathway. Mol Med Rep 2017; 16:2721-2725. [PMID: 28713889 PMCID: PMC5547915 DOI: 10.3892/mmr.2017.6930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/15/2017] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to investigate the role of transcription factor E3 (TFE3) in the regulation of proliferation in renal adenocarcinoma cells. The LV-TFE3 overexpression (OE) lentivirus and negative control CON195 (NC) lentivirus were transfected into the ACHN cell line. Protein expression of FLAG-tag TFE3 was determined using western blot analysis. Differences in cell proliferation, plate clone formation and cell cycle distribution between OE and NC groups were compared using MTT, plate colony formation and flow cytometry assays, respectively. The levels of mammalian target of rapamycin (mTOR) and phosphorylated ribosomal protein S6 (p-rpS6) were analyzed by western blotting. Cell proliferation and colony formation increased significantly in the OE group compared with the NC group. The % of cells in the G1 and G2 phases of the cell cycle decreased, while the % of cells in the S phase of the cell cycle increased in the OE group compared with the NC group. In addition, mTOR and p-rpS6 levels were increased in the OE group compared with the NC group. The results of the present study demonstrated that TFE3 overexpression resulted in increased ACHN cell proliferation and plate clone formation. TFE3 may promote renal tumor growth by regulating cell cycle progression and activating the phosphatidylinositol 3-kinase/AKT serine/threonine kinase 1/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuan Fang
- Reproductive Medical Center, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Bao
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiuyuan Xia
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qin Shen
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiaojun Zhou
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Bing Yao
- Reproductive Medical Center, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
83
|
Kakoki K, Miyata Y, Mochizuki Y, Iwata T, Obatake M, Abe K, Nagayasu T, Sakai H. Long-term Treatment With Sequential Molecular Targeted Therapy for Xp11.2 Translocation Renal Cell Carcinoma: A Case Report and Review of the Literature. Clin Genitourin Cancer 2017; 15:e503-e506. [DOI: 10.1016/j.clgc.2016.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/22/2022]
|
84
|
Zhang L, Yu C, Gao R, Niu Y, Li Y, Chen J, He J. An impedimetric biosensor for the diagnosis of renal cell carcinoma based on the interaction between 3-aminophenyl boronic acid and sialic acid. Biosens Bioelectron 2017; 92:434-441. [DOI: 10.1016/j.bios.2016.10.083] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
|
85
|
RBM10-TFE3 Renal Cell Carcinoma: A Potential Diagnostic Pitfall Due to Cryptic Intrachromosomal Xp11.2 Inversion Resulting in False-negative TFE3 FISH. Am J Surg Pathol 2017; 41:655-662. [PMID: 28296677 DOI: 10.1097/pas.0000000000000835] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Xp11 translocation renal cell carcinoma (RCC) are defined by chromosome translocations involving the Xp11 breakpoint which results in one of a variety of TFE3 gene fusions. TFE3 break-apart florescence in situ hybridization (FISH) assays are generally preferred to TFE3 immunohistochemistry (IHC) as a means of confirming the diagnosis in archival material, as FISH is less sensitive to the variable fixation which can result in false positive or false negative IHC. Prompted by a case report in the cytogenetics literature, we identify 3 cases of Xp11 translocation RCC characterized by a subtle chromosomal inversion involving the short arm of the X chromosome, resulting in an RBM10-TFE3 gene fusion. TFE3 rearrangement was not detected by conventional TFE3 break-apart FISH, but was suggested by strong diffuse TFE3 immunoreactivity in a clean background. We then developed novel fosmid probes to detect the RBM10-TFE3 gene fusion in archival material. These cases validate RBM10-TFE3 as a recurrent gene fusion in Xp11 translocation RCC, illustrate a source of false-negative TFE3 break-apart FISH, and highlight the complementary role of TFE3 IHC and TFE3 FISH.
Collapse
|
86
|
Marchionni L, Hayashi M, Guida E, Ooki A, Munari E, Jabboure FJ, Dinalankara W, Raza A, Netto GJ, Hoque MO, Argani P. MicroRNA expression profiling of Xp11 renal cell carcinoma. Hum Pathol 2017; 67:18-29. [PMID: 28411178 DOI: 10.1016/j.humpath.2017.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/27/2017] [Accepted: 03/18/2017] [Indexed: 12/21/2022]
Abstract
Renal cell carcinomas (RCCs) with Xp11 translocation (Xp11 RCC) constitute a distinctive molecular subtype characterized by chromosomal translocations involving the Xp11.2 locus, resulting in gene fusions between the TFE3 transcription factor with a second gene (usually ASPSCR1, PRCC, NONO, or SFPQ). RCCs with Xp11 translocations comprise up to 1% to 4% of adult cases, frequently displaying papillary architecture with epithelioid clear cells. To better understand the biology of this molecularly distinct tumor subtype, we analyze the microRNA (miRNA) expression profiles of Xp11 RCC compared with normal renal parenchyma using microarray and quantitative reverse-transcription polymerase chain reaction. We further compare Xp11 RCC with other RCC histologic subtypes using publically available data sets, identifying common and distinctive miRNA signatures along with the associated signaling pathways and biological processes. Overall, Xp11 RCC more closely resembles clear cell rather than papillary RCC. Furthermore, among the most differentially expressed miRNAs specific for Xp11 RCC, we identify miR-148a-3p, miR-221-3p, miR-185-5p, miR-196b-5p, and miR-642a-5p to be up-regulated, whereas miR-133b and miR-658 were down-regulated. Finally, Xp11 RCC is most strongly associated with miRNA expression profiles modulating DNA damage responses, cell cycle progression and apoptosis, and the Hedgehog signaling pathway. In summary, we describe here for the first time the miRNA expression profiles of a molecularly distinct type of renal cancer associated with Xp11.2 translocations involving the TFE3 gene. Our results might help understanding the molecular underpinning of Xp11 RCC, assisting in developing targeted treatments for this disease.
Collapse
Affiliation(s)
- Luigi Marchionni
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Center for Computational Genomics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| | - Masamichi Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Elisa Guida
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Akira Ooki
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Enrico Munari
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pathology, Sacro Cuore Hospital Negrar, Verona 37024, Italy
| | - Fayez J Jabboure
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wikum Dinalankara
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ali Raza
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - George J Netto
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pathology, University of Alabama School of Medicine, Birmingham, AL 35233, USA
| | - Mohammad O Hoque
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pedram Argani
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
87
|
Classe M, Malouf GG, Su X, Yao H, Thompson EJ, Doss DJ, Grégoire V, Lenobin J, Fantoni JC, Sudour-Bonnange H, Khayat D, Aubert S, Tannir NM, Leroy X. Incidence, clinicopathological features and fusion transcript landscape of translocation renal cell carcinomas. Histopathology 2017; 70:1089-1097. [DOI: 10.1111/his.13167] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/16/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Marion Classe
- Département de Pathologie; Hôpital Lariboisière; Assistance Publique Hôpitaux de Paris; Paris France
| | - Gabriel G Malouf
- Département d'Oncologie Médicale; Groupe Hospitalier Pitié-Salpêtrière; Assistance Publique Hôpitaux de Paris; Faculté de Médecine Pierre et Marie Curie; Institut Universitaire de Cancérologie GRC5; Paris France
| | - Xiaoping Su
- Department of Bioinformatics and Computational Biology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Hui Yao
- Department of Bioinformatics and Computational Biology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Erika J Thompson
- Department of Genetics; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - Denaha J Doss
- Department of Genetics; The University of Texas MD Anderson Cancer Center; Houston Texas USA
| | - Valérie Grégoire
- Département de Pathologie; Centre Hospitalier Régional Universitaire; Lille France
| | - Julien Lenobin
- Département d'Urologie; Centre Hospitalier Régional Universitaire; Lille France
| | | | | | - David Khayat
- Département d'Oncologie Médicale; Groupe Hospitalier Pitié-Salpêtrière; Assistance Publique Hôpitaux de Paris; Faculté de Médecine Pierre et Marie Curie; Institut Universitaire de Cancérologie GRC5; Paris France
| | - Sébastien Aubert
- Département de Pathologie; Centre Hospitalier Régional Universitaire; Lille France
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Xavier Leroy
- Département de Pathologie; Centre Hospitalier Régional Universitaire; Lille France
| |
Collapse
|
88
|
El Naili R, Nicolas M, Gorena A, Policarpio-Nicolas MLC. Fine-needle aspiration findings of Xp11 translocation renal cell carcinoma metastatic to a hilar lymph node. Diagn Cytopathol 2017; 45:456-462. [PMID: 28185421 DOI: 10.1002/dc.23676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 12/30/2016] [Accepted: 01/18/2017] [Indexed: 01/11/2023]
Abstract
Xp11 translocation renal cell carcinoma (RCC) is a specific type of renal cell carcinoma recently placed under the "MiT family translocation RCC" at the last 2013 ISUP Vancouver classification of renal neoplasia. This tumor contains variable proportions of clear cells and could easily mimic papillary RCC, clear cell type, and clear cell papillary RCC. Given the small number of published cytologic findings of this tumor, it could easily present as a diagnostic pitfall. We describe a case of a 23-year-old man with a history of prior nephrectomy who presented with multiple mediastinal lymphadenopathies on imaging surveillance follow-up. Fine-needle aspiration of the lymph node showed tumor cells with voluminous clear to eosinophilic cytoplasm, well-defined cell borders and hyperchromatic nuclei arranged in papillary architecture. Review of the prior nephrectomy specimen showed papillary cores surrounded by cells with voluminous clear to finely granular eosinophilic cytoplasm and distinct cell borders. Immunohistochemical stains performed on the nephrectomy specimen showed tumor positivity for CD10, E-cadherin, a-methylacyl coenzyme A racemase, and TFE3 supporting the diagnosis of Xp11 translocation renal cell carcinoma. Although this tumor was initially described predominantly in children, it could also occur in adults, as seen in this case. Familiarity with the cytologic findings of this tumor, use of immunohistochemical stains, or cytogenetic test to determine the type of gene fusion will be extremely useful in arriving at the correct diagnosis. Diagn. Cytopathol. 2017;45:456-462. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Reima El Naili
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Marlo Nicolas
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Amanda Gorena
- Pathology Group of Louisiana, Baton Rouge, Louisiana
| | | |
Collapse
|
89
|
Ellis CL, Harik LR, Cohen C, Osunkoya AO. Biomarker, Molecular, and Technologic Advances in Urologic Pathology, Oncology, and Imaging. Arch Pathol Lab Med 2017; 141:499-516. [PMID: 28157406 DOI: 10.5858/arpa.2016-0263-sa] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Urologic pathology is evolving rapidly. Emerging trends include the expanded diagnostic utility of biomarkers and molecular testing, as well as adapting to the plethora of technical advances occurring in genitourinary oncology, surgical practice, and imaging. We illustrate those trends by highlighting our approach to the diagnostic workup of a few selected disease entities that pathologists may encounter, including newly recognized subtypes of renal cell carcinoma, pheochromocytoma, and prostate cancer, some of which harbor a distinctive chromosomal translocation, gene loss, or mutation. We illustrate applications of immunohistochemistry for differential diagnosis of needle core renal biopsies, intraductal carcinoma of the prostate, and amyloidosis and cite encouraging results from early studies using targeted gene expression panels to predict recurrence after prostate cancer surgery. At our institution, pathologists are working closely with urologic surgeons and interventional radiologists to explore the use of intraoperative frozen sections for margins and nerve sparing during robotic prostatectomy, to pioneer minimally invasive videoscopic inguinal lymphadenectomy, and to refine image-guided needle core biopsies and cryotherapy of prostate cancer as well as blue-light/fluorescence cystoscopy. This collaborative, multidisciplinary approach enhances clinical management and research, and optimizes the care of patients with urologic disorders.
Collapse
Affiliation(s)
| | | | | | - Adeboye O Osunkoya
- From the Departments of Pathology (Drs Ellis, Harik, Cohen, and Osunkoya), Urology (Dr Osunkoya), and the Winship Cancer Institute (Dr Osunkoya), Emory University School of Medicine, Atlanta, Georgia; and the Department of Pathology, Veterans Affairs Medical Center, Atlanta, Georgia (Dr Osunkoya)
| |
Collapse
|
90
|
Abstract
The MiT family of translocation-associated renal cell carcinomas comprise approximately 40 % of renal cell carcinomas in young patients but only up to 4 % of renal cell carcinomas in adult patients. The Xp11.2 translocation-associated tumors are the most frequent and were included in the 2004 World Health Organization (WHO) classification. They contain a fusion of the TFE3 gene with ASPSCR1, PRCC, NONO, SPFQ or CLTC resulting in an immunohistochemically detectable nuclear overexpression of TFE3. The Xp11.2 translocation-associated renal cell carcinomas are characterized by ample clear cytoplasm, papillary architecture and abundant psammoma bodies. The TFEB translocation-associated renal cell carcinomas are much rarer and show a biphasic architecture. Fluorescence in situ hybridization permits the detection of a translocation by means of a break apart probe for the TFE3 and TFEB genes and is recommended for the diagnosis of renal cell carcinomas in patients under 30 years of age. The TFE3 and TFEB translocation-associated tumors are classified as MiT family translocation carcinomas in the new WHO classification.The rare renal cell carcinomas harboring an ALK rearrangement with fusion to VCL in young patients with sickle cell trait show a characteristic morphology and are listed in the new WHO classification as a provisional entity.
Collapse
|
91
|
Brok J, Treger TD, Gooskens SL, van den Heuvel-Eibrink MM, Pritchard-Jones K. Biology and treatment of renal tumours in childhood. Eur J Cancer 2016; 68:179-195. [PMID: 27969569 DOI: 10.1016/j.ejca.2016.09.005] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 08/25/2016] [Accepted: 09/01/2016] [Indexed: 02/08/2023]
Abstract
In Europe, almost 1000 children are diagnosed with a malignant renal tumour each year. The vast majority of cases are nephroblastoma, also known as Wilms' tumour (WT). Most children are treated according to Société Internationale d'Oncologie Pédiatrique Renal Tumour Study Group (SIOP-RTSG) protocols with pre-operative chemotherapy, surgery, and post-operative treatment dependent on stage and histology. Overall survival approaches 90%, but a subgroup of WT, with high-risk histology and/or relapsed disease, still have a much poorer prognosis. Outcome is similarly poor for the rare non-WT, particularly for malignant rhabdoid tumour of the kidney, metastatic clear cell sarcoma of the kidney (CCSK), and metastatic renal cell carcinoma (RCC). Improving outcome and long-term quality of life requires more accurate risk stratification through biological insights. Biomarkers are also needed to signpost potential targeted therapies for high-risk subgroups. Our understanding of Wilms' tumourigenesis is evolving and several signalling pathways, microRNA processing and epigenetics are now known to play pivotal roles. Most rhabdoid tumours display somatic and/or germline mutations in the SMARCB1 gene, whereas CCSK and paediatric RCC reveal a more varied genetic basis, including characteristic translocations. Conducting early-phase trials of targeted therapies is challenging due to the scarcity of patients with refractory or relapsed disease, the rapid progression of relapse and the genetic heterogeneity of the tumours with a low prevalence of individual somatic mutations. A further consideration in improving population survival rates is the geographical variation in outcomes across Europe. This review provides a comprehensive overview of the current biological knowledge of childhood renal tumours alongside the progress achieved through international collaboration. Ongoing collaboration is needed to ensure consistency of outcomes through standardised diagnostics and treatment and incorporation of biomarker research. Together, these objectives constitute the rationale for the forthcoming SIOP-RTSG 'UMBRELLA' study.
Collapse
Affiliation(s)
- Jesper Brok
- Cancer Section, University College London, Institute of Child Health, UK; Department of Paediatric Haematology and Oncology, Rigshospitalet, Copenhagen University Hospital, Denmark.
| | - Taryn D Treger
- Cancer Section, University College London, Institute of Child Health, UK
| | - Saskia L Gooskens
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands; Department of Paediatric Haematology and Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Marry M van den Heuvel-Eibrink
- Department of Paediatric Oncology, Princess Máxima Center for Pediatric Oncology and University of Utrecht, The Netherlands
| | | |
Collapse
|
92
|
Calcagnì A, Kors L, Verschuren E, De Cegli R, Zampelli N, Nusco E, Confalonieri S, Bertalot G, Pece S, Settembre C, Malouf GG, Leemans JC, de Heer E, Salvatore M, Peters DJ, Di Fiore PP, Ballabio A. Modelling TFE renal cell carcinoma in mice reveals a critical role of WNT signaling. eLife 2016; 5. [PMID: 27668431 PMCID: PMC5036965 DOI: 10.7554/elife.17047] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT β-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.
Collapse
Affiliation(s)
- Alessia Calcagnì
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Lotte Kors
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Pathology, Academical Medical Center, Amsterdam, The Netherlands
| | - Eric Verschuren
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Nicolina Zampelli
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Edoardo Nusco
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Stefano Confalonieri
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy
| | - Salvatore Pece
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, United States.,Medical Genetics, Federico II University, Naples, Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy
| | - Gabriel G Malouf
- Department of Medical Oncology Groupe Hospitalier Pitie-Salpetriere, University Paris 6, Paris, France.,Assistance Publique Hopitaux de Paris, University Paris 6, Paris, France.,Faculty of Medicine Pierre et Marie Curie, University Paris 6, Paris, France.,Institut Universitaire de Cancerologie GRC5, University Paris 6, Paris, France
| | - Jaklien C Leemans
- Department of Pathology, Academical Medical Center, Amsterdam, The Netherlands
| | - Emile de Heer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Dorien Jm Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Pier Paolo Di Fiore
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, United States.,Medical Genetics, Federico II University, Naples, Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy
| |
Collapse
|
93
|
Ishiguro N, Yoshida H. ASPL-TFE3 Oncoprotein Regulates Cell Cycle Progression and Induces Cellular Senescence by Up-Regulating p21. Neoplasia 2016; 18:626-635. [PMID: 27673450 PMCID: PMC5037204 DOI: 10.1016/j.neo.2016.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 08/05/2016] [Accepted: 08/11/2016] [Indexed: 01/30/2023] Open
Abstract
Alveolar soft part sarcoma is an extremely rare soft tissue sarcoma with poor prognosis. It is characterized by the unbalanced recurrent chromosomal translocation der(17)t(X;17)(p11;q25), resulting in the generation of an ASPL-TFE3 fusion gene. ASPL-TFE3 oncoprotein functions as an aberrant transcriptional factor and is considered to play a crucial role in the tumorigenesis of alveolar soft part sarcoma. However, the underlying molecular mechanisms are poorly understood. In this study, we identified p21 (p21WAF1/CIP1) as a direct transcriptional target of ASPL-TFE3. Ectopic ASPL-TFE3 expression in 293 cells resulted in cell cycle arrest and significant increases in protein and mRNA levels of p21. ASPL-TFE3 activated p21 expression in a p53-independent manner through direct transcriptional interactions with the p21 promoter region. When ASPL-TFE3 was expressed in human bone marrow–derived mesenchymal stem cells in a tetracycline-inducible manner, we observed the up-regulation of p21 expression and the induction of senescence-associated β-galactosidase activity. Suppression of p21 significantly decreased the induction of ASPL-TFE3-mediated cellular senescence. Furthermore, ASPL-TFE3 expression in mesenchymal stem cells resulted in a significant up-regulation of proinflammatory cytokines associated with senescence-associated secretory phenotype (SASP). These results show that ASPL-TFE3 regulates cell cycle progression and induces cellular senescence by up-regulating p21 expression. In addition, our data suggest a potential mechanism by which ASPL-TFE3-induced senescence may play a role in tumorigenesis by inducing SASP, which could promote the protumorigenic microenvironment.
Collapse
Affiliation(s)
- Naoko Ishiguro
- Department of Pathobiological Science and Technology, Faculty of Medicine, Tottori University, 86 Nishimachi, Yonago, Tottori 683-8503, Japan.
| | - Haruhiko Yoshida
- Department of Pathology, Yonago Medical Center, 4-17-1 Kuzumo, Yonago, Tottori 683-0006, Japan
| |
Collapse
|
94
|
Hirsch MS, Signoretti S, Dal Cin P. Adult Renal Cell Carcinoma: A Review of Established Entities from Morphology to Molecular Genetics. Surg Pathol Clin 2016; 8:587-621. [PMID: 26612217 DOI: 10.1016/j.path.2015.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
According to the current World Health Organization (WHO), renal cell carcinomas (RCCs) that primarily affect adults are classified into 8 major subtypes. Additional emerging entities in renal neoplasia have also been recently recognized and these are discussed in further detail by Mehra et al (Emerging Entities in Renal Neoplasia, Surgical Pathology Clinics, 2015, Volume 8, Issue 4). In most cases, the diagnosis of a RCC subtype can be based on morphologic criteria, but in some circumstances the use of ancillary studies can aid in the diagnosis. This review discusses the morphologic, genetic, and molecular findings in RCCs previously recognized by the WHO, and provides clues to distinction from each other and some of the newer subtypes of RCC. As prognosis and therapeutic options vary for the different subtypes of RCC, accurate pathologic distinction is critical for patient care.
Collapse
Affiliation(s)
- Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Paola Dal Cin
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| |
Collapse
|
95
|
Ciccarese C, Di Nunno V, Montironi R, Fiorentino M, Brunelli M, Tortora G, Ardizzoni A, Massari F. The role of precision medicine for the treatment of metastatic renal cell carcinoma. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1214057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Vincenzo Di Nunno
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Sant’Orsola-Malpighi Hospital, Bologna, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata (AOUI), University of Verona, Verona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Andrea Ardizzoni
- Division of Oncology, S.Orsola-Malpighi Hospital, Bologna, Italy
| | | |
Collapse
|
96
|
Ciccarese C, Brunelli M, Montironi R, Fiorentino M, Iacovelli R, Heng D, Tortora G, Massari F. The prospect of precision therapy for renal cell carcinoma. Cancer Treat Rev 2016; 49:37-44. [PMID: 27453294 DOI: 10.1016/j.ctrv.2016.07.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 02/01/2023]
Abstract
The therapeutic landscape of renal cell carcinoma (RCC) has greatly expanded in the last decade. From being a malignancy orphan of effective therapies, kidney cancer has become today a tumor with several treatment options. Renal cell carcinoma (RCC) is a metabolic disease, being characterized by the dysregulation of metabolic pathways involved in oxygen sensing (VHL/HIF pathway alterations and the subsequent up-regulation of HIF-responsive genes such as VEGF, PDGF, EGF, and glucose transporters GLUT1 and GLUT4, which justify the RCC reliance on aerobic glycolysis), energy sensing (fumarate hydratase-deficient, succinate dehydrogenase-deficient RCC, mutations of HGF/MET pathway resulting in the metabolic Warburg shift marked by RCC increased dependence on aerobic glycolysis and the pentose phosphate shunt, augmented lipogenesis, and reduced AMPK and Krebs cycle activity) and/or nutrient sensing cascade (deregulation of AMPK-TSC1/2-mTOR and PI3K-Akt-mTOR pathways). In this complex scenario it is important to find prognostic and predictive factors that can help in decision making in the treatment of mRCC.
Collapse
Affiliation(s)
- Chiara Ciccarese
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostic, Azienda Ospedaliera Universitaria Integrata (AOUI), University of Verona, Verona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Michelangelo Fiorentino
- Pathology Service, Addarii Institute of Oncology, S-Orsola-Malpighi Hospital, Bologna, Italy
| | - Roberto Iacovelli
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Daniel Heng
- Department of Medical Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| | - Giampaolo Tortora
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | | |
Collapse
|
97
|
Chen X, Zhu Q, Li B, Cui W, Zhou H, Duan N, Liu Y, Kundra V, Wang Z. Renal cell carcinoma associated with Xp11.2 translocation/TFE gene fusion: imaging findings in 21 patients. Eur Radiol 2016; 27:543-552. [DOI: 10.1007/s00330-016-4421-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 02/29/2016] [Accepted: 05/19/2016] [Indexed: 02/03/2023]
|
98
|
Wang XT, Xia QY, Ni H, Wang ZY, Ye SB, Li R, Wang X, Lv JH, Shi SS, Ma HH, Lu ZF, Shen Q, Zhou XJ, Rao Q. Xp11 neoplasm with melanocytic differentiation of the prostate harbouring the novel NONO-TFE3 gene fusion: report of a unique case expanding the gene fusion spectrum. Histopathology 2016; 69:450-8. [PMID: 26844676 DOI: 10.1111/his.12949] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/16/2022]
Abstract
Recently, an increasing number of TFE3 rearrangement-associated tumours have been reported, such as TFE3 rearrangement-associated perivascular epithelioid cell tumours (PEComas), melanotic Xp11 translocation renal cancers and melanotic Xp11 neoplasms. We have suggested that these tumours belong to a single clinicopathological spectrum. 'Xp11 neoplasm with melanocytic differentiation' or 'melanotic Xp11 neoplasm' have been proposed to designate this unique neoplasm. Herein, we describe the first case of an Xp11 neoplasm with melanocytic differentiation to be described in the prostate, bearing the novel NONO-TFE3 gene fusion. This study both adds to the spectrum regarding melanotic Xp11 neoplasms and expands its gene fusion spectrum. Moreover, we discuss the relationship of these rare tumours to neoplasms such as conventional PEComas, alveolar soft part sarcomas, malignant melanomas, clear cell sarcomas and Xp11 translocation renal cancers.
Collapse
Affiliation(s)
- Xiao-Tong Wang
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.,Department of Pathology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiu-Yuan Xia
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Hao Ni
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.,Department of Pathology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zi-Yu Wang
- School of Basic Medical Sciences, Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Sheng-Bing Ye
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Rui Li
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Xuan Wang
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jing-Huan Lv
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Shan-Shan Shi
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Heng-Hui Ma
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Zhen-Feng Lu
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Qin Shen
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Xiao-Jun Zhou
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Qiu Rao
- Department of Pathology, Nanjing Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China.,Department of Pathology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
99
|
Magers MJ, Udager AM, Mehra R. MiT Family Translocation-Associated Renal Cell Carcinoma: A Contemporary Update With Emphasis on Morphologic, Immunophenotypic, and Molecular Mimics. Arch Pathol Lab Med 2016; 139:1224-33. [PMID: 26414466 DOI: 10.5858/arpa.2015-0196-ra] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Translocation-associated renal cell carcinoma (t-RCC) is a relatively uncommon subtype of renal cell carcinoma characterized by recurrent gene rearrangements involving the TFE3 or TFEB loci. TFE3 and TFEB are members of the microphthalmia transcription factor (MiT) family, which regulates differentiation in melanocytes and osteoclasts, and MiT family gene fusions activate unique molecular programs that can be detected immunohistochemically. Although the overall clinical behavior of t-RCC is variable, emerging molecular data suggest the possibility of targeted approaches to advanced disease. Thus, distinguishing t-RCC from its morphologic, immunophenotypic, and molecular mimics may have important clinical implications. The differential diagnosis for t-RCC includes a variety of common renal neoplasms, particularly those demonstrating clear cell and papillary features; in addition, because of immunophenotypic overlap and/or shared molecular abnormalities (ie, TFE3 gene rearrangement), a distinctive set of nonepithelial renal tumors may also warrant consideration. Directed ancillary testing is an essential aspect to the workup of t-RCC cases and may include a panel of immunohistochemical stains, such as PAX8, pancytokeratins, epithelial membrane antigen, carbonic anhydrase IX, HMB-45, and Melan-A. Dual-color, break-apart fluorescent in situ hybridization for TFE3 or TFEB gene rearrangement may be helpful in diagnostically challenging cases or when molecular confirmation is needed.
Collapse
Affiliation(s)
- Martin J Magers
- From the Department of Pathology (Drs Magers, Udager, and Mehra), and the Comprehensive Cancer Center (Dr Mehra), University of Michigan Health System, Ann Arbor; and the Michigan Center for Translational Pathology, Ann Arbor (Dr Mehra)
| | | | | |
Collapse
|
100
|
D'Antonio A, Addesso M, Nappi O, Zeppa P. Unsuspected Xp11 Translocation Renal Neoplasm Associated With Contralateral Clear Cell Carcinoma. Int J Surg Pathol 2016; 24:248-52. [PMID: 26729550 DOI: 10.1177/1066896915597753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this report, we present for the first time the coexistence of a conventional renal cell carcinoma (RCC) and an undefined Xp11 translocation renal neoplasm in distinct kidneys, which was difficult to definitively classify as either carcinoma or PEComa (perivascular epithelioid cell tumor). While one of the tumors showed the morphological and immunohistochemical features of clear RCC, the other had an unusual morphology with a prominent nested pattern. Microscopically this tumor showed nests of cells with clear and eosinophilic cytoplasm and nuclei with prominent nucleoli; some hyaline globules were evident. Immunohistochemical panel showed negativity for cytokeratin-pan, cytokeratin-7, PAX8, and CD10 but positive immunostaining for cathepsin K, racemase, Melan-A, and TFE3. A subsequent, metaphase, dual-color fluorescence in situ hybridization confirmed the Xp11 translocation. Attention should be paid to the routine immunohistochemical profile that, in case of negativity of specific RCC markers, may suggest an Xp11 translocation renal tumor. The addition of TFE3 can easily identify the specific subtype.
Collapse
Affiliation(s)
| | | | | | - Pio Zeppa
- University of Medicine and Surgery, Salerno, Italy
| |
Collapse
|