51
|
Tsivian E, Tsivian M, Sze C, Schulman A, Polascik TJ. Clinicopathological characteristics of surgically treated localized renal masses in patients previously exposed to chemotherapy. Int Braz J Urol 2019; 45:332-339. [PMID: 30676301 PMCID: PMC6541126 DOI: 10.1590/s1677-5538.ibju.2018.0126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 10/22/2018] [Indexed: 12/18/2022] Open
Abstract
Purpose: To explore the potential association between renal mass characteristics and a history of chemotherapy. Materials and methods: A retrospective review of records of patients surgically treated for a localized renal mass between 2000 and 2012 was undertaken following an institutional review board approval. Patients age and sex, renal mass clinical characteristics (radiological size and mode of presentation) and pathological characteristics (diagnosis, renal cell carcinoma subtype, Fuhrman grade and stage) were compared between patients with and without a history of chemotherapy, using Fisher's exact test, Student's t-test and Wilcoxon rank sum test. A multivariate logistic analysis was performed to evaluate the independent association of chemotherapy and tumor pathology. Results: Of the 1,038 eligible patients, 33 (3%) had a history of chemotherapy. The distribution of clinical stage, renal mass diagnosis, renal cell carcinoma subtype, Fuhrman grade, pathological stage, sex and median age were similar between the general population and the chemotherapy group. However, the latter had a higher rate of incidental presentation (P = 0.003) and a significantly smaller median radiological tumor size (P = 0.01). In a subset analysis of T1a renal cell carcinoma, the chemotherapy group presented an increased rate of high Fuhrman grade (P = 0.03). On multivariate analysis adjusted for radiological tumor size, sex and age the chemotherapy cohort had a 3.92 higher odds for high Fuhrman grade. Conclusion: Patients with a history of chemotherapy typically present with smaller renal masses that, if malignant, have higher odds of harboring a high Fuhrman grade and thus may not be suitable for active surveillance.
Collapse
Affiliation(s)
- Efrat Tsivian
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, USA
| | - Matvey Tsivian
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, USA
| | - Christina Sze
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, USA
| | - Ariel Schulman
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, USA
| | - Thomas J Polascik
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, USA
| |
Collapse
|
52
|
Harris WB. Epidemiology of Renal Cell Carcinoma and Its Predisposing Risk Factors. Urol Oncol 2019. [DOI: 10.1007/978-3-319-42623-5_55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
53
|
Chang JC, Zhang L, Drilon AE, Chi P, Alaggio R, Borsu L, Benayed R, Travis WD, Ladanyi M, Antonescu CR. Expanding the Molecular Characterization of Thoracic Inflammatory Myofibroblastic Tumors beyond ALK Gene Rearrangements. J Thorac Oncol 2018; 14:825-834. [PMID: 30550870 DOI: 10.1016/j.jtho.2018.12.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/28/2018] [Accepted: 12/02/2018] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Half of inflammatory myofibroblastic tumors (IMTs) regardless of anatomic location harbor anaplastic lymphoma kinase gene (ALK) rearrangements and overexpress anaplastic lymphoma kinase protein. The wide application of next-generation sequencing and the clinical benefit to tyrosine kinase inhibitors have opened new opportunities for investigation of ALK-negative IMTs. METHODS In this study, we have investigated a series of pediatric and adult thoracic IMTs for abnormalities in a wide spectrum of actionable kinases by applying a variety of molecular and next-generation sequencing techniques, including fluorescence in situ hybridization (FISH), targeted RNA sequencing, and NanoString assay. RESULTS There were 33 patients with thoracic IMTs, including five children; their mean age was 37. The tumors showed a monomorphic spindle cell phenotype, except for one with an epithelioid morphologic pattern and moderate to severe atypia. By immunohistochemistry, 24 tumors were ALK positive, and 19 of the 24 showed ALK rearrangements and one ret proto-oncogene gene (RET) rearrangement by FISH. RNA sequencing was performed in the remaining four cases lacking ALK abnormalities by FISH, revealing ALK fusions involving tropomyosin 4 gene (TMP4) and echinoderm microtubule associated protein like 4 gene (EML4) as partner in three cases. NanoString assay was performed in the remaining case, revealing ALK alternative transcription initiation (ALKATI). Nine cases lacking ALK abnormalities were further tested by FISH or targeted RNA sequencing, revealing ROS1 rearrangement in six cases and ETS variant 6 gene (ETV6)-neurotrophic receptor tyrosine kinase 3 gene (NTRK3) fusion in three cases, respectively. CONCLUSIONS By using a battery of complementary molecular techniques, we have shown that all the thoracic IMTs harbored a tyrosine kinase abnormality, with 30% involving a kinase gene other than ALK, including ROS1, NTRK3, and RET gene fusions. We have also described for the first time ALKATI-induced ALK oncogenic activation in IMTs.
Collapse
Affiliation(s)
- Jason C Chang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lei Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander E Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ping Chi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rita Alaggio
- Department of Pathology, University Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Laetitia Borsu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ryma Benayed
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Travis
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina R Antonescu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
54
|
Signoretti S, Flaifel A, Chen YB, Reuter VE. Renal Cell Carcinoma in the Era of Precision Medicine: From Molecular Pathology to Tissue-Based Biomarkers. J Clin Oncol 2018; 36:JCO2018792259. [PMID: 30372384 PMCID: PMC6299340 DOI: 10.1200/jco.2018.79.2259] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is not a single entity but includes various tumor subtypes that have been identified on the basis of either characteristic pathologic features or distinctive molecular changes. Clear cell RCC is the most common type of RCC and is characterized by dysregulation of the von Hippel Lindau/hypoxia-inducible factor pathway. Non-clear cell RCC represents a more heterogeneous group of tumors with diverse histopathologic and molecular features. In the past two decades, the improved understanding of the molecular landscape of RCC has led to the development of more effective therapies for metastatic RCC, which include both targeted agents and immune checkpoint inhibitors. Because only subsets of patients with metastatic RCC respond to a given treatment, predictive biomarkers are needed to guide treatment selection and sequence. In this review, we describe the key histologic features and molecular alterations of RCC subtypes and discuss emerging tissue-based biomarkers of response to currently available therapies for metastatic disease.
Collapse
Affiliation(s)
- Sabina Signoretti
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Abdallah Flaifel
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Ying-Bei Chen
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Victor E. Reuter
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| |
Collapse
|
55
|
Saad AM, Gad MM, Al-Husseini MJ, Ruhban IA, Sonbol MB, Ho TH. Trends in Renal-Cell Carcinoma Incidence and Mortality in the United States in the Last 2 Decades: A SEER-Based Study. Clin Genitourin Cancer 2018; 17:46-57.e5. [PMID: 30391138 DOI: 10.1016/j.clgc.2018.10.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 09/17/2018] [Accepted: 10/04/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Renal-cell carcinoma (RCC) is one of the common malignancies in the United States. RCC incidence and mortality have been changing for many reasons. We performed a thorough investigation of incidence and mortality trends of RCC in the United States using the cell Surveillance, Epidemiology, and End Results (SEER) database. PATIENTS AND METHODS The 13 SEER registries were accessed for RCC cases diagnosed between 1992 and 2015. Incidence and mortality were calculated by demographic and tumor characteristics. We calculated annual percentage changes of these rates. Rates were expressed as 100,000 person-years. RESULTS A total of 104,584 RCC cases were reviewed, with 47,561 deaths. The overall incidence was 11.281 per 100,000 person-years. Incidence increased by 2.421% per year (95% confidence interval, 2.096, 2.747; P < .001) but later became stable since 2008. However, the incidence of clear-cell subtype continued to increase (1.449%; 95% confidence interval, 0.216, 2.697; P = .024). RCC overall mortality rates have been declining since 2001. However, mortality associated with distant RCC only started to decrease in 2012, with an annual percentage change of 18.270% (95% confidence interval, -28.775, -6.215; P = .006). CONCLUSION Despite an overall increase in the incidence of RCC, there has been a recent plateau in RCC incidence rates with a significant decrease in mortality.
Collapse
Affiliation(s)
- Anas M Saad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mohamed M Gad
- Faculty of Medicine, Ain Shams University, Cairo, Egypt; Heart and Vascular Institute, Cleveland Clinic Foundation, Cleveland, OH
| | | | - Inas A Ruhban
- Pathology department, Faculty of Medicine, Damascus University, Damascus, Syria
| | | | - Thai H Ho
- Mayo Clinic Cancer Center, Phoenix, AZ.
| |
Collapse
|
56
|
Palsgrove DN, Li Y, Lin MT, Pallavajjalla A, Gocke C, De Marzo AM, Matoso A, Netto GJ, Epstein JI, Argani P. Eosinophilic Solid and Cystic (ESC) Renal Cell Carcinomas Harbor TSC Mutations: Molecular Analysis Supports an Expanding Clinicopathologic Spectrum. Am J Surg Pathol 2018; 42:1166-1181. [PMID: 29975249 PMCID: PMC6089659 DOI: 10.1097/pas.0000000000001111] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Eosinophilic solid and cystic (ESC) renal cell carcinoma (RCC) has recently been described as a potentially new subtype of RCC based upon morphologic and immunohistochemical features. These neoplasms typically demonstrate solid and cystic architecture, and the neoplastic cells contain voluminous eosinophilic cytoplasm with granular cytoplasmic stippling. There is frequently focal immunoreactivity for cytokeratin 20. Although the initial cases all occurred in adult females and had benign outcome, we recently expanded the proposed spectrum of this neoplasm to include pediatric cases, multifocal neoplasms, and a case with hematogenous metastasis. ESC has been postulated to be analogous to a subtype of RCC consistently identified in tuberous sclerosis complex patients, and while previous work has demonstrated loss of heterozygosity at the TSC1 locus and copy number gains at TSC2 in ESC RCC, these genes have not been sequenced in ESC RCC. Using capture-based and amplicon-based next-generation sequencing, we now demonstrate the consistent presence of either TSC1 or TSC2 gene mutations in pediatric ESC RCC (8/9 cases) and adult ESC RCC (6/6 cases). These included a metastatic ESC RCC which had a complete response to mTOR targeted therapy. We also found these mutations in some neoplasms with variant morphology and thus potentially expand the spectrum of ESC RCC. These include one of our adult cases which demonstrated dominant "type 2" papillary RCC morphology and 2 of 3 previously unclassified pediatric RCC with features of ESC RCC minus granular cytoplasmic stippling. We also demonstrate TSC mutations in a case of so-called "oncocytoid RCC after neuroblastoma" with identical morphology and immunoprofile, providing a molecular link between the latter and ESC RCC. In summary, ESC RCC consistently harbors actionable TSC1 or TSC2 mutations, which are infrequently seen in established subtypes of RCC. These findings support TSC1/2 mutation as a molecular marker of ESC RCC, and suggest expansion of the clinicopathologic spectrum to include neoplasms with papillary architecture, occasional cases lacking well-developed granular cytoplasmic stippling, and a subset of RCC with oncocytic features in patients who have survived neuroblastoma.
Collapse
Affiliation(s)
- Doreen N. Palsgrove
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Yunjie Li
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Ming-Tseh Lin
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Aparna Pallavajjalla
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Christopher Gocke
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Angelo M. De Marzo
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Andres Matoso
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - George J. Netto
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Jonathan I. Epstein
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Pedram Argani
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| |
Collapse
|
57
|
Ma J, Pan C, Yin M. Translocation Renal Cell Carcinoma in a Child Previously Treated for Infantile Fibrosarcoma. Pediatr Dev Pathol 2018; 21:418-422. [PMID: 28478739 DOI: 10.1177/1093526617707849] [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] [Indexed: 11/15/2022]
Abstract
We report a child who developed a TFE3/Xp11.2 translocation renal cell carcinoma (RCC) when he was 3 years and 3 months old, after previous treatment for infantile fibrosarcoma (IFS). When he was 3 months old, a left axilla mass has been detected, which was tan and solid, was 1.5 cm in greatest dimension, and composed of sheets of spindle cells that was positive for vimentin and fibronectin. Fluorescence in situ hybridization showed positive result in ETV6 gene rearrangements. The final diagnosis was IFS. After surgery and chemotherapy, he remained disease-free until 3 years; later, he was detected to have a tumor in right kidney which measured 2.5 × 2 × 1.5 cm. The tumor comprised clear-cell features that were arranged in papillary and adenoid architecture. The tumor cells were positive for TFE3 and CK. The diagnosis was TFE3/Xp11.2 translocation RCC. Previous research has reported that the radio/chemotherapy for the first tumor might be involved in the pathogenesis of translocation RCC. In our report, this is the first time the IFS is included in the disease spectrum which can cause secondary translocation RCC.
Collapse
Affiliation(s)
- Jing Ma
- 1 Department of Pathology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ci Pan
- 2 Department of Hematology and Oncology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minzhi Yin
- 1 Department of Pathology, Shanghai Children's Medical Center Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
58
|
Li Y, Reuter VE, Matoso A, Netto GJ, Epstein JI, Argani P. Re-evaluation of 33 'unclassified' eosinophilic renal cell carcinomas in young patients. Histopathology 2018; 72:588-600. [PMID: 28898443 PMCID: PMC7582203 DOI: 10.1111/his.13395] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 09/07/2017] [Indexed: 12/16/2022]
Abstract
AIMS We sought to determine if some unclassified renal cell carcinomas (RCCs) in children and young adults that are characterised by predominantly eosinophilic cytoplasm are related to the recently described succinate dehydrogenase (SDH)-deficient RCC, fumarate hydratase (FH)-deficient RCC or eosinophilic solid and cystic (ESC) RCC. METHODS AND RESULTS We reviewed 33 unclassified RCCs with predominantly eosinophilic cytoplasm in patients aged 35 years or younger. Immunohistochemistry (IHC) for SDHB, FH and CK20 (a marker of ESC) was performed in all cases. IHC for 2-succinocysteine (2SC) was performed on RCC with loss of FH labelling. Four RCC (12%) (median age 18 years) demonstrated loss of FH labelling as well as aberrant 2SC labelling, and were thus classified as FH-deficient RCCs. Importantly, none of these cases demonstrated the characteristic macronucleoli typical of FH-deficient RCC. Eight RCC (24%) (median age 20.5 years) demonstrated loss of SDHB and were reclassified as SDH-deficient RCCs. Importantly, only four of eight SDH-deficient RCC demonstrated the characteristic cytoplasmic vacuoles and inclusions of typical SDH-deficient RCC. Ten RCC (30%) (median age 27 years) were reclassified as ESC RCCs. Four of 10 ESC RCC were multifocal (one bilateral), four of 10 ESC RCC occurred in males and one patient presented with liver and lung metastases, all not described previously in ESC. Eleven RCC (33%) remained unclassified. CONCLUSIONS Pathologists should have a low threshold for performing FH, SDHB and CK20 IHC when confronted with unclassified eosinophilic RCC or 'oncocytoma' in young patients.
Collapse
Affiliation(s)
- Yunjie Li
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Victor E Reuter
- Memorial Sloan Kettering Cancer Center, Pathology, New York, NY, United States
| | - Andres Matoso
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - George J Netto
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
- University of Alabama at Birmingham School of Medicine, Birmingham,AL, USA
| | - Jonathan I Epstein
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| | - Pedram Argani
- Johns Hopkins University School of Medicine, Pathology, Baltimore, MD, United States
| |
Collapse
|
59
|
Caliò A, Brunelli M, Segala D, Pedron S, Tardanico R, Remo A, Gobbo S, Meneghelli E, Doglioni C, Hes O, Zampini C, Argani P, Martignoni G. t(6;11) renal cell carcinoma: a study of seven cases including two with aggressive behavior, and utility of CD68 (PG-M1) in the differential diagnosis with pure epithelioid PEComa/epithelioid angiomyolipoma. Mod Pathol 2018; 31:474-487. [PMID: 29052596 DOI: 10.1038/modpathol.2017.144] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 01/21/2023]
Abstract
Renal cell carcinomas with t(6;11) chromosome translocation involving the TFEB gene are indolent neoplasms which often occur in young patients. In this study, we report seven cases of renal cell carcinoma with TFEB rearrangement, two of whom had histologically proven metastasis. Patients (4F, 3M) ranged in age from 19 to 55 years (mean 37). One patient developed paratracheal and pleural metastases 24 months after surgery and died of disease after 46 months; another one recurred with neoplastic nodules in the perinephric fat and pelvic soft tissue. Histologically, either cytological or architectural appearance was peculiar in each case whereas one tumor displayed the typical biphasic morphology. By immunohistochemistry, all tumors labelled for cathepsin K, Melan-A and CD68 (KP1 clone). HMB45 and PAX8 staining were detected in six of seven tumors. All tumors were negative for CD68 (PG-M1 clone), CKAE1-AE3, CK7, CAIX, and AMACR. Seven pure epithelioid PEComa/epithelioid angiomyolipomas, used as control, were positive for cathepsin K, melanocytic markers, and CD68 (PG-M1 and KP1) and negative for PAX8. Fluorescence in situ hybridization results showed the presence of TFEB gene translocation in all t(6;11) renal cell carcinomas with a high frequency of split TFEB fluorescent signals (mean 74%). In the primary and metastatic samples of the two aggressive tumors, increased gene copy number was observed (3-5 fluorescent signals per neoplastic nuclei) with a concomitant increased number of CEP6. Review of the literature revealed older age and larger tumor size as correlating with aggressive behavior in these neoplasms. In conclusion, we present the clinical, morphological and molecular features of seven t(6;11) renal cell carcinomas, two with histologically demonstrated metastasis. We report the high frequency of split signals by FISH in tumors with t(6;11) chromosomal rearrangement and the occurrence of TFEB gene copy number gains in the aggressive cases, analyzing either the primary or metastatic tumor. Finally, we demonstrate the usefulness of CD68 (PG-M1) immunohistochemical staining in distinguishing t(6;11) renal cell carcinoma from pure epithelioid PEComa/epithelioid angiomyolipoma.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Diego Segala
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Serena Pedron
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | | | - Andrea Remo
- Department of Pathology, Hospital 'Mater Salutis', Legnago, Italy
| | - Stefano Gobbo
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Emanuela Meneghelli
- Department of Life and Reproduction Sciences, Clinical Biochemistry Laboratory, University of Verona, Verona, Italy
| | | | - Ondrej Hes
- Department of Pathology, Charles University Hospital Plzen, Plzen, Czech Republic
| | - Claudia Zampini
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Pedram Argani
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, USA
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy.,Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| |
Collapse
|
60
|
Zhan HQ, Li ST, Shu Y, Liu MM, Qin R, Li YL, Gan L. Alpha gene upregulates TFEB expression in renal cell carcinoma with t(6;11) translocation, which promotes cell canceration. Int J Oncol 2018; 52:933-944. [DOI: 10.3892/ijo.2018.4239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/20/2017] [Indexed: 11/06/2022] Open
Affiliation(s)
- He-qin Zhan
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shu-ting Li
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan Shu
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Meng-meng Liu
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Rong Qin
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yan-li Li
- Department of Pathology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Lin Gan
- Institute of Clinical Virology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
61
|
Chung EM, Lattin GE, Fagen KE, Kim AM, Pavio MA, Fehringer AJ, Conran RM. Renal Tumors of Childhood: Radiologic-Pathologic Correlation Part 2. The 2nd Decade: From the Radiologic Pathology Archives. Radiographics 2017; 37:1538-1558. [PMID: 28898190 DOI: 10.1148/rg.2017160189] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malignant renal tumors account for 7% of childhood cancers, and Wilms tumors are by far the most common-but not in older children and adolescents. Among individuals in the latter half of their 2nd decade of life, renal cell carcinoma (RCC) is more common than Wilms tumor. The histopathologic spectrum of RCCs in children differs from that in adults. The most common subtype of RCC in children and adolescents is Xp11.2 translocation RCC, which is distinguished by hyperattenuation at nonenhanced computed tomography, a defined capsule, and associated retroperitoneal lymphadenopathy. Papillary RCC is the second most common histologic subtype. It enhances less intensely compared with the adjacent renal parenchyma and has a propensity for calcification. Clear cell RCC is seen in patients with von Hippel-Lindau disease and is distinguished by its relatively hypervascular nature. Medullary carcinoma affects adolescents with the sickle cell trait and is characterized by an infiltrative growth pattern and extensive metastasis at presentation. Angiomyolipoma is seen in children with tuberous sclerosis complex and is often multifocal and hypervascular, with macroscopic fat. Metanephric tumors are central, circumscribed, and typically calcified. Lymphoma usually manifests as multifocal masses, but it may involve a solitary mass or infiltrative pattern. Extensive adenopathy and involvement of the gastrointestinal tract or other organs also may be seen. Primitive neuroectodermal tumor is an aggressive neoplasm that is typically quite large at diagnosis. Knowledge of the clinical, biologic, and histopathologic features of renal tumors in older children and adolescents and their effects on the imaging appearance can help the radiologist offer a useful preoperative differential diagnosis.
Collapse
Affiliation(s)
- Ellen M Chung
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Grant E Lattin
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Kimberly E Fagen
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Andrew M Kim
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Michael A Pavio
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Adam J Fehringer
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| | - Richard M Conran
- From the Department of Radiology and Radiological Sciences, F. Edward Hébert School of Medicine (A.J.F.), Uniformed Services University of the Health Sciences, 4301 Jones Bridge Rd, Bethesda, MD 20814 (E.M.C., G.E.L.); Pediatric Radiology Section (E.M.C.) and Genitourinary Radiology Section (G.E.L.), American Institute for Radiologic Pathology, Silver Spring, Md; Department of Radiology (K.E.F.) and Office of Graduate Medical Education (A.M.K., M.A.P.), Walter Reed National Military Medical Center, Bethesda, Md; and Department of Pathology, Eastern Virginia Medical School, Norfolk, Va (R.M.C.)
| |
Collapse
|
62
|
Hypoxia induces TFE3 expression in head and neck squamous cell carcinoma. Oncotarget 2017; 7:11651-63. [PMID: 26872381 PMCID: PMC4905500 DOI: 10.18632/oncotarget.7309] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/23/2016] [Indexed: 12/16/2022] Open
Abstract
To assess the role of transcription factor μE3 (TFE3) in the tumorigenesis of head and neck squamous cell carcinoma (HNSCC), human HNSCC tissue arrays were investigated for TFE3 expression. Human HNSCC tissues with neoadjuvant inductive chemotherapey (docetaxel, cisplatin and fluorouracil, TPF) and mice HNSCC tissues from transgenic mice model were evaluated for TFE3 expression and the hypoxia pathway. The roles of EGF/EGFR mediated hypoxia in TFE3 nuclear expression were analyzed in vitro and in vivo. TFE3 expression was higher in human HNSCC tissues compared with that in normal oral mucosa. Moreover, high TFE3 expression was related to HIF-1α, PAI-1, and EGFR, which demonstrated the activation of the hypoxia pathway in HNSCC tissues. Furthermore, elevated TFE3 expression was observed in HNSCC after cisplatin-based chemotherapy, and high TFE3 expression may indicate poor response to TPF inductive chemotherapy. Furthermore, similar changes with increased TFE3 were observed in HNSCC of the transgenic mouse HNSCC model. Hypoxic culture in the human HNSCC cell line increased TFE3 expression, which promoted cell survival under hypoxia. EGFR inhibiton by cetuximab could attenuate hypoxia-induced TFE3 in the HNSCC cell line and transgenic mouse HNSCC model. These findings indicated that TFE3 was an important hypoxia-induced transcriptional factor in HNSCC. TFE3 could be regarded as a durgable therapeutic oncotarget by EGFR inhibition.
Collapse
|
63
|
Rhee DS, Dunkel IJ, Shukla NN, Walsh MF, Gilheeney SW, Price AP, Antonescu CR, Heaton TE. Alveolar soft part sarcoma of the bladder with ASPSCR1-TFE3 gene fusion as a secondary malignancy. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2017. [DOI: 10.1016/j.epsc.2017.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
64
|
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: 34] [Impact Index Per Article: 4.9] [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
|
65
|
Renal Cell Carcinoma Occurring in Patients With Prior Neuroblastoma: A Heterogenous Group of Neoplasms. Am J Surg Pathol 2017; 40:989-97. [PMID: 26975037 DOI: 10.1097/pas.0000000000000632] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Renal cell carcinoma (RCC) associated with neuroblastoma (NB) was included as a distinct entity in the 2004 World Health Organization classification of kidney tumors. A spectrum of RCC subtypes has been reported in NB survivors. We herein describe a series of 8 RCCs diagnosed in 7 patients with a history of NB. Microscopic evaluation, immunohistochemical staining for PAX8, cathepsin K, and succinate dehydrogenase subunit B (SDHB), and fluorescence in situ hybridization (FISH) for TFE3 and TFEB were performed. Four distinct morphologic subtypes were identified: 3 tumors were characterized by cells with abundant oncocytoid cytoplasm and irregular nuclei; 3 showed features of microphthalmia transcription factor family translocation RCC (MiTF-RCC); 1 had features of hybrid oncocytic-chromophobe tumor; 1 had papillary RCC histology. All RCCs expressed PAX8 and retained SDHB expression. Cathepsin K was positive in 2 MiTF-RCCs, 1 was TFEB FISH positive, and the other was indeterminate. Cathepsin K was negative in a third MiTF-RCC with TFE3 rearrangement. TFE3 FISH was negative in 4 and insufficient in 1 of the other 5 RCCs. While a subset of RCCs associated with NB is characterized by cells with prominent oncocytoid cytoplasm, other RCC subtypes also occur in post-NB patients. Renal neoplasms occurring in patients with a history of NB do not represent a single entity but a heterogenous group of RCCs. SDHB mutations do not explain the subset of nontranslocation RCCs with oncocytoid features; therefore, further studies are needed to clarify whether they may represent a distinct entity with unique molecular abnormalities or may belong to other emerging RCC subtypes.
Collapse
|
66
|
Schaefer BA, Johnson TS, Hooper DK, Nathan JD, Geller JI. TFE3-positive renal cell carcinoma occurring in three children with dysfunctional kidneys on immunosuppression. Pediatr Transplant 2017; 21. [PMID: 28322484 DOI: 10.1111/petr.12912] [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] [Accepted: 02/06/2017] [Indexed: 12/20/2022]
Abstract
Pediatric RCC is a rare pediatric neoplasm and is distinctly different compared to adult RCC, often demonstrating translocation morphology evidenced by unique histopathological features and TFE3 or TFEB nuclear expression. We report three cases of pediatric TFE3 positive RCC (TFE3-RCC) occurring in the setting of chronic kidney disease and long-term pharmacological immunosuppression, including two cases that developed in the native kidney following kidney transplantation. Together, these cases suggest that the kidney microenvironment in combination with immune dysregulation is likely contributing factors in the pathogenesis of some pediatric RCC, warranting further study. Long-term post-transplant surveillance may warrant screening for RCC.
Collapse
Affiliation(s)
- Beverly A Schaefer
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Theodore S Johnson
- Division of Pediatric Hematology/Oncology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David K Hooper
- Division of Pediatric Nephrology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Jaimie D Nathan
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - James I Geller
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
67
|
Choo MS, Jeong CW, Song C, Jeon HG, Seo SI, Hong SK, Byun SS, Chung JS, Hong SH, Hwang EC, Kim HH, Kwak C. Clinicopathologic Characteristics and Prognosis of Xp11.2 Translocation Renal Cell Carcinoma: Multicenter, Propensity Score Matching Analysis. Clin Genitourin Cancer 2017; 15:e819-e825. [PMID: 28549862 DOI: 10.1016/j.clgc.2017.04.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/08/2017] [Accepted: 04/14/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND We evaluated the clinicopathologic characteristics and prognosis of Xp11.2 translocation (Xp11.2t) renal cell carcinoma (RCC) from a multicenter study and compare them with clear-cell RCC using a propensity score matching analysis. PATIENTS AND METHODS Between 2004 and 2013, 8384 consecutive patients from 7 institutions who were diagnosed with RCC were reviewed, and the pathologically confirmed Xp11.2t cases were enrolled. The oncological outcomes of Xp11.2t were compared with those of clear-cell RCC by selecting matched cases using 1:3 propensity score matching methods in a precollected clear-cell RCC data set from our hospital. The patients were divided into 2 subgroups on the basis of age of onset, either before (early) or after (late) 45 years old. RESULTS Xp11.2t was found in 61 cases, corresponding to 0.72% of RCC cases for the 10 years. The mean age was 38.2 ± 19.4 years, and the mean tumor size was 6.2 ± 3.9 cm. The Xp11.2t cases were at more advanced stages and showed tendencies to involve lymph nodes at diagnosis. After the matching, there were no significant differences in recurrence-free and overall survival compared with clear-cell RCC. The age of incidence for Xp11.2t had a bimodal distribution, which was most common in the 30s and smaller peak in the 60s. Xp11.2t corresponded to a significantly worse prognosis for overall survival in late onset (after 45 years) subgroup (P = .038; hazard ratio, 3.199; 95% confidence interval, 1.065-9.609). CONCLUSION This neoplasm has more aggressive clinicopathologic features at diagnosis. In older patients with onset age > 45 years, Xp11.2t showed a significantly worse prognosis than clear-cell RCC.
Collapse
Affiliation(s)
- Min Soo Choo
- Department of Urology, Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Chang Wook Jeong
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Cheryn Song
- Department of Urology, Asan Medical Center, Seoul, Korea
| | - Hwang Gyun Jeon
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seong Il Seo
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung Kyu Hong
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jin Soo Chung
- Department of Urology, National Cancer Center, Goyang, Korea
| | - Sung-Hoo Hong
- Department of Urology, Kangnam St Mary's Hospital, Seoul, Korea
| | - Eu Chang Hwang
- Department of Urology, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Hyeon Hoe Kim
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University Hospital, Seoul, Korea.
| | | |
Collapse
|
68
|
Udager AM, Mehra R. Morphologic, Molecular, and Taxonomic Evolution of Renal Cell Carcinoma: A Conceptual Perspective With Emphasis on Updates to the 2016 World Health Organization Classification. Arch Pathol Lab Med 2017; 140:1026-37. [PMID: 27684973 DOI: 10.5858/arpa.2016-0218-ra] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Molecular and morphologic interrogation has driven a much-needed reexamination of renal cell carcinoma (RCC). Indeed, the recently released 2016 World Health Organization classification now recognizes 12 distinct RCC subtypes, as well as several other emerging/provisional RCC entities. From a clinical perspective, accurate RCC classification may have important implications for patients and their families, including prognostic risk stratification, targeted therapeutics selection, and identification for genetic testing. In this review, we provide a conceptual framework for approaching RCC diagnosis and classification by categorizing RCCs as tumors with clear cytoplasm, papillary architecture, and eosinophilic (oncocytic) cytoplasm. The currently recognized 2016 World Health Organization classification for RCC subtypes is briefly discussed, including new diagnostic entities (clear cell papillary RCC, hereditary leiomyomatosis and RCC-associated RCC, succinate dehydrogenase-deficient RCC, tubulocystic RCC, and acquired cystic disease-associated RCC) and areas of evolving RCC classification, such as transcription elongation factor B subunit 1 (TCEB1)-mutated RCC/RCC with angioleiomyoma-like stroma/RCC with leiomyomatous stroma, RCC associated with anaplastic lymphoma receptor tyrosine kinase (ALK) gene rearrangement, thyroidlike follicular RCC, and RCC in neuroblastoma survivors. For each RCC subtype, relevant clinical, molecular, gross, and microscopic findings are reviewed, and ancillary studies helpful for its differential diagnosis are presented, providing a practical approach to modern RCC classification.
Collapse
Affiliation(s)
- Aaron M Udager
- From the Department of Pathology (Drs 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
|
69
|
Abstract
The fourth edition of the World Health Organization (WHO) report was published in January 2016 and focused especially on the kidney tumors and several new concepts. Many steps forward in the subclassification have been taken. Several new entities have been added, as major progress on a genetic level has been made. Some already well-known entities have new prognosis in accordance with longer follow up and research, and some concepts of well-known groups have been refined.
Collapse
Affiliation(s)
- Eva Compérat
- Service d'anatomie et cytologie pathologique, Hôpital Tenon, UPMC Paris VI, Paris.
| | - Justine Varinot
- Service d'anatomie et cytologie pathologique, Hôpital Tenon, UPMC Paris VI, Paris
| |
Collapse
|
70
|
Ellati RT, Abukhiran I, Alqasem K, Jasser J, Khzouz J, Bisharat T, Al-saidi I, Al-Daghmin A. Clinicopathologic Features of Translocation Renal Cell Carcinoma. Clin Genitourin Cancer 2017; 15:112-116. [DOI: 10.1016/j.clgc.2016.05.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 01/28/2023]
|
71
|
[WHO classification 2016 and first S3 guidelines on renal cell cancer: What is important for the practice?]. DER PATHOLOGE 2017; 37:127-33. [PMID: 26942728 DOI: 10.1007/s00292-016-0144-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The first S3 guidelines on renal cell cancer cover the practical aspects of imaging, diagnostics and therapy as well as the clinical relevance of pathology reporting. This review summarizes the changes in renal tumor classification and the new recommendations for reporting renal cell tumors. The S3 guidelines recommend the 2016 World Health Organization (WHO) classification of renal cell tumors. Novel renal cell tumor entities and provisional or emerging renal cell tumor entities of the 2016 WHO classification of renal tumors are discussed. The S3 guidelines for renal cell cancer also recommend the use of the WHO/International Society of Urologic Pathology (ISUP) grading system for clear cell and for papillary renal cell carcinomas, which replaces the previously used Fuhrman grading system.
Collapse
|
72
|
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
|
73
|
Bezhanova SD. [Tumors of the kidney. The new 2016 WHO classification of tumors of the genitourinary system]. Arkh Patol 2017; 79:48-52. [PMID: 28418358 DOI: 10.17116/patol201779248-52] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The article deals with the innovations available in the 2016 World Health Organization (WHO) classification of renal tumors, which has replaced the previous 2004 WHO classification. The review highlights the key aspects necessary for practicing histopathologists who work in specialized cancer centers or multidisciplinary hospitals diagnosing renal tumors. How to choose a system for determining the malignancy grade of renal tumors is considered separately.
Collapse
Affiliation(s)
- S D Bezhanova
- N.N. Blokhin Russian Cancer Research Center, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
74
|
Epidemiology of Renal Cell Carcinoma and Its Predisposing Risk Factors. Urol Oncol 2017. [DOI: 10.1007/978-3-319-42603-7_55-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
75
|
Oh MJ, Jeong JH, Im SB, Kwak JJ, Nam KH. Neural Axis Metastasis from Metachronous Pulmonary Basaloid Carcinoma Developed after Chemotherapy & Radiation Therapy of Uterine Cervical Carcinoma. Korean J Neurotrauma 2016; 12:167-170. [PMID: 27857930 PMCID: PMC5110911 DOI: 10.13004/kjnt.2016.12.2.167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/29/2016] [Accepted: 04/27/2016] [Indexed: 11/15/2022] Open
Abstract
Multiple primary or secondary malignancies after anticancer therapy were recently reported to be increasing in frequency. The authors describe a case of metachronous metastatic pulmonary basaloid carcinoma to the central nervous system that was discovered after chemotherapy and radiation therapy for cervical uterine carcinoma. Two different types of cancer developed within some interval. There's the possibility that a secondary pulmonary neoplasm developed after the chemotherapy and radiotherapy conducted as cervical cancer treatment.
Collapse
Affiliation(s)
- Myeong Jin Oh
- Department of Neurosurgery, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Je Hoon Jeong
- Department of Neurosurgery, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Soo Bin Im
- Department of Neurosurgery, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Jeong Ja Kwak
- Department of Pathology, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| | - Kye-Hyun Nam
- Department of Obstetrics and Gynecology, Soonchunhyang University College of Medicine, Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
76
|
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: 96] [Impact Index Per Article: 12.0] [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
|
77
|
Thorner PS, Shago M, Marrano P, Shaikh F, Somers GR. TFE3-positive renal cell carcinomas are not always Xp11 translocation carcinomas: Report of a case with a TPM3-ALK translocation. Pathol Res Pract 2016; 212:937-942. [PMID: 27450657 DOI: 10.1016/j.prp.2016.07.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/08/2016] [Indexed: 01/16/2023]
Abstract
Translocation-associated renal cell carcinoma (RCC) is a distinct subtype of RCC with gene rearrangements of the TFE3 or TFEB loci. The TFE3 gene is located at Xp11 and can fuse to a number of translocation partners, resulting in high nuclear expression of TFE3 protein. TFE3 immunostaining is often used as a surrogate marker for a TFE3 translocation. We report a case of an RCC that expressed TFE3 but showed only gain of TFE3 rather than a translocation. Moreover, this case had a t(1;2) translocation fusing ALK and TMP3, identical to that seen in inflammatory myofibroblastic tumour. There was resulting overexpression of ALK protein in a cytoplasmic and membranous pattern. The patient was not treated with chemotherapy but following regional nodal recurrence, an ALK inhibitor was added and the patient remains alive one year later. There are only rare reports of RCC with an ALK-TMP3 fusion, and these tumours can express TFE3 on some unknown basis not related to a TFE3 translocation. Any RCC positive for TFE3 and lacking a translocation should be tested for ALK expression and translocation. Recognition of this subtype of RCC will allow ALK inhibitor therapy to be added, in the hope of improving patient outcome.
Collapse
Affiliation(s)
- Paul Scott Thorner
- Division of Pathology, Hospital for Sick Children, Toronto, Canada; Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, Canada.
| | - Mary Shago
- Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, Canada; Division of Genome Diagnostics, Hospital for Sick Children, Toronto, Canada
| | - Paula Marrano
- Division of Pathology, Hospital for Sick Children, Toronto, Canada
| | - Furqan Shaikh
- Division of Hematology/Oncology, Hospital for Sick Children, Toronto, Canada; Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Gino R Somers
- Division of Pathology, Hospital for Sick Children, Toronto, Canada; Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|
78
|
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: 1.1] [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]
|
79
|
Chauhan NS, Bharti R, Chander B, Kumar S. Pediatric Clear Cell Renal Cell Carcinoma with Pelvicalyceal System Invasion and Contiguous Extension Upto Bladder: Novel and Bizarre Tumor Behaviour. Pol J Radiol 2016; 81:256-60. [PMID: 27298654 PMCID: PMC4886614 DOI: 10.12659/pjr.896088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/13/2015] [Indexed: 11/09/2022] Open
Abstract
Background Paediatric renal cell carcinoma (RCC) is a rare neoplasm which differs significantly in its clinico-pathological behaviour from the adult variant. The clear cell variant constitutes a relatively small histological subset of this neoplasm. Case Report We present a very unusual, pathologically proven case of clear cell variety of pediatric RCC which showed invasion into the pelvicalyceal system with contiguous extension up to the urinary bladder. Such a novel manifestation of paediatric RCC has not been described previously in literature. A relevant review of literature is included. Conclusions The aggressive biological behaviour of the paediatric RCC in our case and the consequent atypical imaging findings are distinctly unusual. These findings may represent a new aggressive variant of this rare neoplasm. The radiologist and clinician should be aware of these novel manifestations of paediatric clear cell RCC.
Collapse
Affiliation(s)
- Narvir S Chauhan
- Department of Radiology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Ramesh Bharti
- Department of Surgery, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Bal Chander
- Department of Pathology, Dr. Rajendra Prasad Government Medical College, Kangra, India
| | - Satish Kumar
- Department of Surgery, Dr. Rajendra Prasad Government Medical College, Kangra, India
| |
Collapse
|
80
|
Rao Q, Xia QY, Cheng L, Zhou XJ. Molecular genetics and immunohistochemistry characterization of uncommon and recently described renal cell carcinomas. Chin J Cancer Res 2016; 28:29-49. [PMID: 27041925 DOI: 10.3978/j.issn.1000-9604.2016.01.03] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) compromises multiple types and has been emerging dramatically over the recent several decades. Advances and consensus have been achieved targeting common RCCs, such as clear cell carcinoma, papillary RCC and chromophobe RCC. Nevertheless, little is known on the characteristics of several newly-identified RCCs, including clear cell (tubulo) papillary RCC, Xp11 translocation RCC, t(6;11) RCC, succinate dehydrogenase (SDH)-deficient RCC, acquired cystic disease-associated RCC, hereditary leiomyomatosis RCC syndrome-associated RCC, ALK translocation RCC, thyroid-like follicular RCC, tubulocystic RCC and hybrid oncocytic/chromophobe tumors (HOCT). In current review, we will collect available literature of these newly-described RCCs, analyze their clinical pathologic characteristics, discuss their morphologic and immunohistologic features, and finally summarize their molecular and genetic evidences. We expect this review would be beneficial for the understanding of RCCs, and eventually promote clinical management strategies.
Collapse
Affiliation(s)
- Qiu Rao
- 1 Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China ; 2 Department of Pathology and Laboratory, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Qiu-Yuan Xia
- 1 Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China ; 2 Department of Pathology and Laboratory, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Liang Cheng
- 1 Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China ; 2 Department of Pathology and Laboratory, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Jun Zhou
- 1 Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China ; 2 Department of Pathology and Laboratory, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
81
|
LIU JIAJU, SU ZHENGMING, LI YIFAN, CHEN DUQUN, NI LIANGCHAO, MAO XIANGMING, YANG SHANGQI, LAI YONGQING. Xp11.2 translocation renal cell carcinoma with multiple bone metastases: A case report. Oncol Lett 2016; 11:2233-2236. [PMID: 26998154 PMCID: PMC4774413 DOI: 10.3892/ol.2016.4211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 12/18/2015] [Indexed: 02/05/2023] Open
Abstract
Xp11.2 translocation/transcription factor enhancer 3 (TFE3) fusion gene associated with renal cell carcinoma (Xp11.2 translocation RCC) is rare and occurs predominantly in children and adolescents. The current study reports the case of a 14-year-old male with Xp11.2 translocation RCC, who presented with chest pain that had persisted for 1 month. A solid neoplasm was located in the left kidney of the patient. Contrast-enhanced computed tomography revealed the presence of a solid mass in the kidney, with uneven enhancement. Destruction of multiple bones was also observed. The patient was treated with a radical nephrectomy. The pathological examination of the tumor revealed that the tumor cells contained an eosinophilic cytoplasm in the renal interstitial tissue. Immunohistochemistry revealed that the tumor cells expressed P504S, cluster of differentiation 10, pan-cytokeratin, vimentin and TFE3. In conclusion, Xp11.2 translocation RCC is a rare type of kidney cancer. Diagnosing this disease prior to surgery is challenging, and providing a definite diagnosis requires histopathological and immunohistochemical examination, while genetic analysis may also be required.
Collapse
Affiliation(s)
- JIAJU LIU
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Department of Urology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - ZHENGMING SU
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Department of Urology, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - YIFAN LI
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Department of Urology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - DUQUN CHEN
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Department of Urology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - LIANGCHAO NI
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - XIANGMING MAO
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - SHANGQI YANG
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
| | - YONGQING LAI
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, P.R. China
- The Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Peking University Shenzhen Hospital, Institute of Urology of Shenzhen PKU-HKUST Medical Center, Shenzhen, Guangdong 518036, P.R. China
- Correspondence to: Professor Yongqing Lai, Department of Urology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, Guangdong 518036, P.R. China, E-mail:
| |
Collapse
|
82
|
Moch H, Cubilla AL, Humphrey PA, Reuter VE, Ulbright TM. The 2016 WHO Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol 2016; 70:93-105. [PMID: 26935559 DOI: 10.1016/j.eururo.2016.02.029] [Citation(s) in RCA: 1939] [Impact Index Per Article: 242.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
UNLABELLED The fourth edition of the World Health Organization (WHO) classification of urogenital tumours (WHO "blue book"), published in 2016, contains significant revisions. These revisions were performed after consideration by a large international group of pathologists with special expertise in this area. A subgroup of these persons met at the WHO Consensus Conference in Zurich, Switzerland, in 2015 to finalize the revisions. This review summarizes the most significant differences between the newly published classification and the prior version for renal, penile, and testicular tumours. Newly recognized epithelial renal tumours are hereditary leiomyomatosis and renal cell carcinoma (RCC) syndrome-associated RCC, succinate dehydrogenase-deficient RCC, tubulocystic RCC, acquired cystic disease-associated RCC, and clear cell papillary RCC. The WHO/International Society of Urological Pathology renal tumour grading system was recommended, and the definition of renal papillary adenoma was modified. The new WHO classification of penile squamous cell carcinomas is based on the presence of human papillomavirus and defines histologic subtypes accordingly. Germ cell neoplasia in situ (GCNIS) of the testis is the WHO-recommended term for precursor lesions of invasive germ cell tumours, and testicular germ cell tumours are now separated into two fundamentally different groups: those derived from GCNIS and those unrelated to GCNIS. Spermatocytic seminoma has been designated as a spermatocytic tumour and placed within the group of non-GCNIS-related tumours in the 2016 WHO classification. PATIENT SUMMARY The 2016 World Health Organization (WHO) classification contains new renal tumour entities. The classification of penile squamous cell carcinomas is based on the presence of human papillomavirus. Germ cell neoplasia in situ of the testis is the WHO-recommended term for precursor lesions of invasive germ cell tumours.
Collapse
Affiliation(s)
- Holger Moch
- Department of Pathology, University Hospital Zurich, Zurich, Switzerland.
| | - Antonio L Cubilla
- Instituto de Patología e Investigación, Facultad de Ciencias Médicas, Universidad Nacional de Asunción, San Lorenzo, Paraguay
| | - Peter A Humphrey
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas M Ulbright
- Department of Pathology and Laboratory Medicine, Indiana University Health Partners, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
83
|
|
84
|
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: 5.9] [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
|
85
|
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
|
86
|
Young EE, Brown CT, Merguerian PA, Akhavan A. Pediatric and adolescent renal cell carcinoma. Urol Oncol 2016; 34:42-9. [DOI: 10.1016/j.urolonc.2015.06.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/16/2015] [Accepted: 06/16/2015] [Indexed: 12/13/2022]
|
87
|
Fedhila Ben Ayed F, Rhayem S, Doghri R, Ben Hassine L, Khemiri M, Mrad K, Bellagha I, Barsaoui S. [Secondary TFE3-associated renal cell carcinoma in a child treated for Ewing sarcoma]. Arch Pediatr 2015; 23:171-5. [PMID: 26702489 DOI: 10.1016/j.arcped.2015.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/12/2015] [Accepted: 11/09/2015] [Indexed: 11/30/2022]
Abstract
Renal cell carcinoma is a rare pediatric malignant tumor of the kidney. Unlike Wilms tumor, the efficacy of chemotherapy and radiation therapy in pediatric renal cell carcinoma remains uncertain. Surgery is the best treatment and prognosis is favorable when the tumor is localized and completely eradicated. We report an exceptional observation in a 7-year-old girl with renal cell carcinoma who had been treated 20 months previously for Ewing sarcoma with chemotherapy and radiotherapy. The renal tumor was revealed by abdominal pain without hematuria. She underwent a radical nephrectomy, and histopathology concluded in renal carcinoma associated with translocation Xp 11.2 grade 3 of Furhrman pT3a N1. No adjuvant therapy was given. After 3 years of follow-up, there is no evidence of local or metastatic recurrence. This observation is significant given the very young age of this patient, the occurrence after Ewing sarcoma with a short disease-free interval. It seems that translocation renal cell carcinoma is associated with previous exposure to chemotherapy, particularly topoisomerase II inhibitors or alkylating agents.
Collapse
Affiliation(s)
- F Fedhila Ben Ayed
- Service de médecine infantile A, hôpital d'enfants de Tunis, Bechir Hamza, Bab Saadoun place, 1007 Tunis Jabbari, Tunisie.
| | - S Rhayem
- Service de médecine infantile A, hôpital d'enfants de Tunis, Bechir Hamza, Bab Saadoun place, 1007 Tunis Jabbari, Tunisie
| | - R Doghri
- Service d'anatomopathologie, institut Salah Azaiz, Tunis, Tunisie
| | - L Ben Hassine
- Service de radiologie pédiatrique, hôpital d'enfants de Tunis, Tunis, Tunisie
| | - M Khemiri
- Service de médecine infantile A, hôpital d'enfants de Tunis, Bechir Hamza, Bab Saadoun place, 1007 Tunis Jabbari, Tunisie
| | - K Mrad
- Service d'anatomopathologie, institut Salah Azaiz, Tunis, Tunisie
| | - I Bellagha
- Service de radiologie pédiatrique, hôpital d'enfants de Tunis, Tunis, Tunisie
| | - S Barsaoui
- Service de médecine infantile A, hôpital d'enfants de Tunis, Bechir Hamza, Bab Saadoun place, 1007 Tunis Jabbari, Tunisie
| |
Collapse
|
88
|
PSF/SFPQ is a very common gene fusion partner in TFE3 rearrangement-associated perivascular epithelioid cell tumors (PEComas) and melanotic Xp11 translocation renal cancers: clinicopathologic, immunohistochemical, and molecular characteristics suggesting classification as a distinct entity. Am J Surg Pathol 2015; 39:1181-96. [PMID: 26274027 DOI: 10.1097/pas.0000000000000502] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
An increasing number of TFE3 rearrangement-associated tumors, such as TFE3 rearrangement-associated perivascular epithelioid cell tumors (PEComas), melanotic Xp11 translocation renal cancers, and melanotic Xp11 neoplasms, have recently been reported. We examined 12 such cases, including 5 TFE3 rearrangement-associated PEComas located in the pancreas, cervix, or pelvis and 7 melanotic Xp11 translocation renal cancers, using clinicopathologic, immunohistochemical, and molecular analyses. All the tumors shared a similar morphology, including a purely nested or sheet-like architecture separated by a delicate vascular network, purely epithelioid cells displaying a clear or granular eosinophilic cytoplasm, a lack of papillary structures and spindle cell or fat components, uniform round or oval nuclei containing small visible nucleoli, and, in most cases (11/12), melanin pigmentation. The levels of mitotic activity and necrosis varied. All 12 cases displayed moderately (2+) or strongly (3+) positive immunoreactivity for TFE3 and cathepsin K. One case labeled focally for HMB45 and Melan-A, whereas the others typically labeled moderately (2+) or strongly (3+) for 1 of these markers. None of the cases were immunoreactive for smooth muscle actin, desmin, CKpan, S100, or PAX8. PSF-TFE3 fusion genes were confirmed by reverse transcription polymerase chain reaction in cases (7/7) in which a novel PSF-TFE3 fusion point was identified. All of the cases displayed TFE3 rearrangement associated with Xp11 translocation. Furthermore, we developed a PSF-TFE3 fusion fluorescence in situ hybridization assay for the detection of the PSF-TFE3 fusion gene and detected it in all 12 cases. Clinical follow-up data were available for 7 patients. Three patients died, and 2 patients (cases 1 and 3) remained alive with no evidence of disease after initial resection. Case 2 experienced recurrence and remained alive with disease. Case 5, a recent case, remained alive with extensive abdominal cavity metastases. Our data suggest that these tumors belong to a single clinicopathologic spectrum and expand the known characteristics of TFE3 rearrangement-associated tumors.
Collapse
|
89
|
Abstract
One of the principal roles of a nephrologist is to closely monitor renal transplant allograft function and promptly evaluate any dysfunction. Renal transplant sonography has a major role in this assessment process given its ability to easily define renal transplant anatomy and surrounding structures. Abnormalities can be extrarenal or involve vascular, parenchymal and urological components of the graft and these can acutely or chronically influence graft function and survival. Procedural guidance as is required during allograft biopsy, as well as routine surveillance and screening for post transplant complications such as malignancy are also important applications of ultrasound in the management of renal transplant recipients. This article outlines key ultrasound findings and applications in renal transplantation from the clinician's perspective.
Collapse
Affiliation(s)
- Khai Gene Leong
- Department of Nephrology Monash Health Clayton Victoria Australia
| | - Peter Coombs
- Monash Imaging C/-Monash HealthClaytonVictoriaAustralia; Department of Medical ImagingRadiation Sciences Monash UniversityClaytonVictoriaAustralia
| | - John Kanellis
- Department of Nephrology Monash HealthClaytonVictoriaAustralia; Centre for Inflammatory DiseasesDepartment of Medicine Monash UniversityClaytonVictoriaAustralia
| |
Collapse
|
90
|
Huang W, Goldfischer M, Babayeva S, Mao Y, Volyanskyy K, Dimitrova N, Fallon JT, Zhong M. Identification of a novel PARP14-TFE3 gene fusion from 10-year-old FFPE tissue by RNA-seq. Genes Chromosomes Cancer 2015; 54:500-505. [PMID: 26032162 DOI: 10.1002/gcc.22261] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/08/2015] [Accepted: 04/09/2015] [Indexed: 12/20/2022] Open
Abstract
Xp11 (TFE3) translocation renal cell carcinoma (RCC) is officially recognized as a distinct subtype of RCC in the 2004 WHO classification. This neoplasm is characterized by several chromosomal translocations between the TFE3-involving Xp11.2 breakpoint and various fusion partners. To date, five partner genes have been identified, that is, PRCC in 1q21, PSF in 1q34, ASPL in 17q25, CLTC in 17q23, and NONO in Xq12; and three additional translocations have been reported with no partner gene being defined: t(X;3)(p11;q23), t(X;10)(p11;q23), and t(X;19)(p11;q13). Here, we report the identification of a novel TFE3 fusion partner, PARP14 in chromosome band3q21. We used RNA-seq on a 10-year-old FFPE (formalin-fixed, paraffin-embedded) tissue sample, which carried t(X;3)(p11;q23) as detected in the original cytogenetic study. The fusion transcript connected the 5'-end of the first two exons of PARP14 to the 3'-end of five exons of TFE3, which was verified by reverse transcription PCR (RT-PCR) and Sanger sequencing. Similar to other TFE3 fusions previously reported, the predicted PARP14-TFE3 product retains the nuclear localization and DNA-binding domains of TFE3. This finding expands the list of TFE3 translocation partner genes and re-emphasizes the essential oncogenic role of TFE3 fusion proteins in this tumor. Our result also clearly demonstrated the feasibility of identifying chromosomal translocation by RNA-seq in clinical FFPE, which are easily accessible and associated with valuable clinical information. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Weihua Huang
- Department of Pathology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Michael Goldfischer
- Department of Pathology, Hackensack University Medical Center, Hackensack, NJ
| | - Sabina Babayeva
- Department of Pathology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Yong Mao
- Clinical Informatics Solutions and Services (CISS), Philips Research North America, Briarcliff Manor, NY
| | - Konstantin Volyanskyy
- Clinical Informatics Solutions and Services (CISS), Philips Research North America, Briarcliff Manor, NY
| | - Nevenka Dimitrova
- Clinical Informatics Solutions and Services (CISS), Philips Research North America, Briarcliff Manor, NY
| | - John T Fallon
- Department of Pathology, New York Medical College, Westchester Medical Center, Valhalla, NY
| | - Minghao Zhong
- Department of Pathology, New York Medical College, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
91
|
Adult Patient with Synchronous Gastrointestinal Stromal Tumor and Xp11 Translocation-Associated Renal Cell Carcinoma: A Unique Case Presentation with Discussion and Review of Literature. Case Rep Urol 2015; 2015:814809. [PMID: 26246933 PMCID: PMC4515533 DOI: 10.1155/2015/814809] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 01/08/2023] Open
Abstract
Gastrointestinal stromal tumor (GIST) is the most common primary mesenchymal tumor of the gastrointestinal tract. This entity comprises a wide spectrum of tumors that vary from benign to overtly malignant, with the majority of these tumors harboring oncogenic mutations of the KIT receptor tyrosine kinase that can aid in diagnosis as well as in targeted therapy. Although the majority of GISTs are sporadic, there are forms that are associated with a variety of syndromes including Carney-Stratakis syndrome and neurofibromatosis type 1, as well as a subset of familial GIST syndromes that are caused by germline mutations in KIT or PDGFRA. Here, we describe an unusual case of a patient who was found to have a large abdominal GIST with an incidentally found Xp11 translocation-associated renal carcinoma. The karyotype of the renal carcinoma revealed an unbalanced rearrangement involving an (X;22) translocation at Xp11.2 and 22p11.2, which has not been reported in the literature. Although GISTs have shown an association with other primary malignant neoplasms, including simultaneous presence with unilateral clear cell renal cell carcinoma and bilateral papillary renal cell carcinomas, we describe the first reported case of synchronous GIST and Xp11 translocation-associated renal cell carcinoma.
Collapse
|
92
|
|
93
|
Abstract
The MiT subfamily of transcription factors includes TFE3, TFEB, TFC, and MiTF. Gene fusions involving two of these transcription factors have been identified in renal cell carcinoma (RCC). The Xp11 translocation RCCs were first officially recognized in the 2004 WHO renal tumor classification, and harbor gene fusions involving TFE3. The t(6;11) RCCs harbor a specific Alpha-TFEB gene fusion and were first officially recognized in the 2013 International Society of Urologic Pathology (ISUP) Vancouver classification of renal neoplasia. These two subtypes of translocation RCC have many similarities. Both were initially described in and disproportionately involve young patients, though adult translocation RCC may overall outnumber pediatric cases. Both often have unusual and distinctive morphologies; the Xp11 translocation RCCs frequently have clear cells with papillary architecture and abundant psammomatous bodies, while the t(6;11) RCCs frequently have a biphasic appearance with both large and small epithelioid cells and nodules of basement membrane material. However, the morphology of these two neoplasms can overlap, with one mimicking the other. Both of these RCCs underexpress epithelial immunohistochemical markers like cytokeratin and epithelial membrane antigen (EMA) relative to most other RCCs. Unlike other RCCs, both frequently express the cysteine protease cathepsin k and often express melanocytic markers like HMB45 and Melan A. Finally, TFE3 and TFEB have overlapping functional activity as these two transcription factors frequently heterodimerize and bind to the same targets. Therefore, on the basis of clinical, morphologic, immunohistochemical, and genetic similarities, the 2013 ISUP Vancouver classification of renal neoplasia grouped these two neoplasms together under the heading of "MiT family translocation RCC." This review summarizes our current knowledge of these recently described RCCs.
Collapse
Affiliation(s)
- Pedram Argani
- Department of Pathology, The Johns Hopkins University School of Medicine, The Johns Hopkins Hospital, 401 North Broadway, Weinberg 2242, Baltimore, Maryland 21231.
| |
Collapse
|
94
|
Arneja SK, Gujar N. Renal cell carcinoma with t(6:11) (p21;q12). A case report highlighting distinctive immunohistologic features of this rare tumor. Int J Surg Case Rep 2014; 7C:16-9. [PMID: 25562596 PMCID: PMC4336393 DOI: 10.1016/j.ijscr.2014.12.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 12/17/2014] [Accepted: 12/17/2014] [Indexed: 02/03/2023] Open
Abstract
Renal cell carcinoma (RCC) with t(6:11) (p21;q12) are extremely rare, fewer than 30 cases have been reported in literature. This study reports a case of 11 year old boy diagnosed with this rare tumor, with the aim of highlighting distinctiveimmuno-histologic features of this tumor otherwise diagnosed by fluorescence in situ hybridization technique. Histologically tumor has a distinct biphasic pattern, with “pseudorosette” formation. Tumor cells show immunopositivity for melanocytic markers, HMB45 and Melan A and are negative for epithelial markers CK and EMA. Knowledge of unique immunohistology is helpful in not misdiagnosing this rare tumor for a non epithelial tumor like angiomyolipoma.
Introduction Renal cell carcinoma (RCC) with t(6:11) (p21;q12) are extremely rare, fewer than 30 cases have been reported in literature. These tumors are characterized by specific chromosomal translocation involving TFEB, as against the more commonly known TFE3 (Xp11.2) translocation associated RCCs. The distinctive immnohistologic features are helpful in enabling a diagnosis of this rare tumor, otherwise diagnosed by fluorescence in situ hybridization assay, specific for detecting TFEB gene rearrangement. Presentation of case Herein, we report a case of this rare tumor in a 11 years old boy, with the objective of highlighting distinctive light microscopic and immuno-phenotypic features of this rare sub-type of translocation associated renal cell carcinoma, otherwise diagnosed by fluorescence in situ hybridization technique. Morphologically tumor showed distinctive biphasic population of cells, large epitheloid cells with voluminous eosinophillic cytoplasm and smaller cells with much lesser amount of cytoplasm and small rounded nuclei. The smaller cells at places clustered around hyaline pink material forming “pseudorosettes”. population. Immunohistochemically both types of tumor cells showed negativity for pan CK (cytokeratin), EMA (epitheleal membrane antigen) and TFE3 (transcription factor E3). HMB 45 (human melanoma black 45) and Melan- A /MART 1 (melanoma antigen recognized by T cells) were moderate to strongly expressed. Discussion On review of literature, most RCCs with t(6;11) translocation have been reported to be negative for pan cytokeratins and EMA. Published literature also shows that the most distinctive immunohistochemical feature of t(6;11) translocation RCC is nuclear staining for TFEB protein. Immunostains for TFE3 have always been negative in the reported cases. It is noteworthy that immunoreactivity for melanocytic markers HMB45 and Melan A and immunonegativity for epithelial markers pan CK and EMA may lead to misdiagnosis of angiomyolipoma to the unwary. Conclusion Knowledge of distinctive morphological and immuno-histochemical features of this tumor can help in establishing a diagnosis of this rare subset of translocation associated RCC on routine hematoxylin and eosin (H and E) staining and immunophenotyping.
Collapse
Affiliation(s)
- Sarabjeet Kaur Arneja
- Department of Surgical Pathology And Cytology, Breach Candy Hospital Trust, 60 A Bhulabhai Desai Road,Mumbai 400026, India.
| | - Neeraj Gujar
- Department of Surgical Pathology And Cytology, Breach Candy Hospital Trust, 60 A Bhulabhai Desai Road,Mumbai 400026, India.
| |
Collapse
|
95
|
Parihar A, Tickoo SK, Kumar S, Arora VK. Xp11 Translocation Renal Cell Carcinoma Morphologically Mimicking Clear Cell–Papillary Renal Cell Carcinoma in an Adult Patient. Int J Surg Pathol 2014; 23:234-7. [DOI: 10.1177/1066896914562280] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Xp11 translocation renal cell carcinoma (RCC) is a relatively rare tumor mainly affecting children and adolescents. It shows significant morphological overlap with the 2 most common adult renal tumors, which are the clear cell (conventional) RCC and papillary RCC. We describe case of a young adult female who presented with right flank pain and abdominal mass. Radiological investigations showed features suggestive of renal cell carcinoma in the right kidney. Histopathological findings while suggestive of Xp11 carcinoma, showed significant overlap with the recently described entity clear cell papillary RCC. TFE3 immunohistochemistry confirmed the tumor to be Xp11 translocation RCC. The patient had an aggressive course with lymph node metastasis. In this report, we discuss differential diagnosis and the diagnostic challenges of Xp11 translocation RCC in adults.
Collapse
Affiliation(s)
| | | | - Sunil Kumar
- University College of Medical Sciences, Delhi, India
| | | |
Collapse
|
96
|
Peckova K, Vanecek T, Martinek P, Spagnolo D, Kuroda N, Brunelli M, Vranic S, Djuricic S, Rotterova P, Daum O, Kokoskova B, Vesela P, Pivovarcikova K, Bauleth K, Dubova M, Kalusova K, Hora M, Michal M, Hes O. Aggressive and nonaggressive translocation t(6;11) renal cell carcinoma: comparative study of 6 cases and review of the literature. Ann Diagn Pathol 2014; 18:351-7. [DOI: 10.1016/j.anndiagpath.2014.10.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/06/2014] [Indexed: 11/25/2022]
|
97
|
Sudour-Bonnange H, Leroy X, Chauvet MP, Classe M, Robin PM, Leblond P. Cutaneous metastases during an aggressive course of Xp11.2 translocation renal cell carcinoma in a teenager. Pediatr Blood Cancer 2014; 61:1698-700. [PMID: 24585574 DOI: 10.1002/pbc.25015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/04/2014] [Indexed: 11/11/2022]
Abstract
We reported a rare case of cutaneous metastases of renal cell carcinoma (RCC) with an Xp11.2 translocation in a 15-year-old female. Clinicians should be aware of the possibility of this uncommon site of metastasis, which can indicate multivisceral dissemination of the disease. We discuss the feasibility and opportunity of treating such a patient with multiple line of tyrosine kinase inhibitor (TKI) targeting vascular endothelial and platelet-derived growth factor receptors.
Collapse
|
98
|
Kauffman EC, Ricketts CJ, Rais-Bahrami S, Yang Y, Merino MJ, Bottaro DP, Srinivasan R, Linehan WM. Molecular genetics and cellular features of TFE3 and TFEB fusion kidney cancers. Nat Rev Urol 2014; 11:465-75. [PMID: 25048860 DOI: 10.1038/nrurol.2014.162] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Despite nearly two decades passing since the discovery of gene fusions involving TFE3 or TFEB in sporadic renal cell carcinoma (RCC), the molecular mechanisms underlying the renal-specific tumorigenesis of these genes remain largely unclear. The recently published findings of The Cancer Genome Atlas Network reported that five of the 416 surveyed clear cell RCC tumours (1.2%) harboured SFPQ-TFE3 fusions, providing further evidence for the importance of gene fusions. A total of five TFE3 gene fusions (PRCC-TFE3, ASPSCR1-TFE3, SFPQ-TFE3, NONO-TFE3, and CLTC-TFE3) and one TFEB gene fusion (MALAT1-TFEB) have been identified in RCC tumours and characterized at the mRNA transcript level. A multitude of molecular pathways well-described in carcinogenesis are regulated in part by TFE3 or TFEB proteins, including activation of TGFβ and ETS transcription factors, E-cadherin expression, CD40L-dependent lymphocyte activation, mTORC1 signalling, insulin-dependent metabolism regulation, folliculin signalling, and retinoblastoma-dependent cell cycle arrest. Determining which pathways are most important to RCC oncogenesis will be critical in discovering the most promising therapeutic targets for this disease.
Collapse
Affiliation(s)
- Eric C Kauffman
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Christopher J Ricketts
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Soroush Rais-Bahrami
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Youfeng Yang
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Maria J Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| | - W Marston Linehan
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Building 10, CRC Room 1-5940, Bethesda, MD 20892, USA
| |
Collapse
|
99
|
Malouf GG, Su X, Yao H, Gao J, Xiong L, He Q, Compérat E, Couturier J, Molinié V, Escudier B, Camparo P, Doss DJ, Thompson EJ, Khayat D, Wood CG, Yu W, Teh BT, Weinstein J, Tannir NM. Next-generation sequencing of translocation renal cell carcinoma reveals novel RNA splicing partners and frequent mutations of chromatin-remodeling genes. Clin Cancer Res 2014; 20:4129-40. [PMID: 24899691 DOI: 10.1158/1078-0432.ccr-13-3036] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE MITF/TFE translocation renal cell carcinoma (TRCC) is a rare subtype of kidney cancer. Its incidence and the genome-wide characterization of its genetic origin have not been fully elucidated. EXPERIMENTAL DESIGN We performed RNA and exome sequencing on an exploratory set of TRCC (n = 7), and validated our findings using The Cancer Genome Atlas (TCGA) clear-cell RCC (ccRCC) dataset (n = 460). RESULTS Using the TCGA dataset, we identified seven TRCC (1.5%) cases and determined their genomic profile. We discovered three novel partners of MITF/TFE (LUC7L3, KHSRP, and KHDRBS2) that are involved in RNA splicing. TRCC displayed a unique gene expression signature as compared with other RCC types, and showed activation of MITF, the transforming growth factor β1 and the PI3K complex targets. Genes differentially spliced between TRCC and other RCC types were enriched for MITF and ID2 targets. Exome sequencing of TRCC revealed a distinct mutational spectrum as compared with ccRCC, with frequent mutations in chromatin-remodeling genes (six of eight cases, three of which were from the TCGA). In two cases, we identified mutations in INO80D, an ATP-dependent chromatin-remodeling gene, previously shown to control the amplitude of the S phase. Knockdown of INO80D decreased cell proliferation in a novel cell line bearing LUC7L3-TFE3 translocation. CONCLUSIONS This genome-wide study defines the incidence of TRCC within a ccRCC-directed project and expands the genomic spectrum of TRCC by identifying novel MITF/TFE partners involved in RNA splicing and frequent mutations in chromatin-remodeling genes.
Collapse
Affiliation(s)
| | - Xiaoping Su
- Bioinformatics and Computational Biology and
| | - Hui Yao
- Bioinformatics and Computational Biology and
| | | | | | | | - Eva Compérat
- Pathology, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique Hopitaux de Paris, Faculty of Medicine Pierre et Marie Curie, Institut Universitaire de Cancérologie GRC5, University Paris 6
| | | | | | - Bernard Escudier
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif
| | | | | | | | | | - Christopher G Wood
- Division of Surgery, Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, Texas; and
| | - Willie Yu
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Bin T Teh
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | | | | |
Collapse
|
100
|
Clinical heterogeneity of Xp11 translocation renal cell carcinoma: impact of fusion subtype, age, and stage. Mod Pathol 2014; 27:875-86. [PMID: 24309327 DOI: 10.1038/modpathol.2013.208] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 09/13/2013] [Accepted: 09/24/2013] [Indexed: 12/16/2022]
Abstract
Xp11 translocation renal cell carcinomas harbor chromosome translocations involving the Xp11 breakpoint, resulting in gene fusions involving the TFE3 gene. The most common subtypes are the ASPSCR1-TFE3 renal cell carcinomas resulting from t(X;17)(p11;q25) translocation, and the PRCC-TFE3 renal cell carcinomas, resulting from t(X;1)(p11;q21) translocation. A formal clinical comparison of these two subtypes of Xp11 translocation renal cell carcinomas has not been performed. We report one new genetically confirmed Xp11 translocation renal cell carcinoma of each type. We also reviewed the literature for all published cases of ASPSCR1-TFE3 and PRCC-TFE3 renal cell carcinomas and contacted all corresponding authors to obtain or update the published follow-up information. Study of two new, unpublished cases, and review of the literature revealed that 8/8 patients who presented with distant metastasis had ASPSCR1-TFE3 renal cell carcinomas, and all but one of these patients either died of disease or had progressive disease. Regional lymph nodes were involved by metastasis in 24 of the 32 ASPSCR1-TFE3 cases in which nodes were resected, compared with 5 of 14 PRCC-TFE3 cases (P=0.02).; however, 11 of 13 evaluable patients with ASPSCR1-TFE3 renal cell carcinomas who presented with N1M0 disease remained disease free. Two PRCC-TFE3 renal cell carcinomas recurred late (at 20 and 30 years, respectively). In multivariate analysis, only older age or advanced stage at presentation (not fusion subtype) predicted death. In conclusion, ASPSCR1-TFE3 renal cell carcinomas are more likely to present at advanced stage (particularly node-positive disease) than are PRCC-TFE3 renal cell carcinomas. Although systemic metastases portend a grim prognosis, regional lymph node involvement does not, at least in short-term follow-up. The tendency for PRCC-TFE3 renal cell carcinomas to recur late warrants long-term follow-up.
Collapse
|