1
|
Asrani K, Amaral A, Woo J, Abadchi SN, Vidotto T, Feng K, Liu HB, Kasbe M, Baba M, Oike Y, Outeda P, Watnick T, Rosenberg AZ, Schmidt LS, Linehan WM, Argani P, Lotan TL. SFPQ-TFE3 gene fusion reciprocally regulates mTORC1 activity and induces lineage plasticity in a novel mouse model of renal tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.21.624702. [PMID: 39605439 PMCID: PMC11601635 DOI: 10.1101/2024.11.21.624702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The MiT/TFE family gene fusion proteins, such as SFPQ-TFE3 , drive both epithelial (eg, translocation renal cell carcinoma, tRCC) and mesenchymal (eg, perivascular epithelioid cell tumor, PEComa) neoplasms with aggressive behavior. However, no prior mouse models for SFPQ-TFE3 -related tumors exist and the mechanisms of lineage plasticity induced by this fusion remain unclear. Here, we demonstrate that constitutive murine renal expression of human SFPQ-TFE3 using Ksp Cadherin-Cre as a driver disrupts kidney development leading to early neonatal renal failure and death. In contrast, post-natal induction of SFPQ-TFE3 in renal tubular epithelial cells using Pax8 ERT-Cre induces infiltrative epithelioid tumors, which morphologically and transcriptionally resemble human PEComas. As seen in MiT/TFE fusion-driven human tumors, SFPQ-TFE3 expression is accompanied by the strong induction of mTORC1 signaling, which is partially amino acid-sensitive and dependent on increased SFPQ-TFE3 -mediated RRAGC/D transcription. Remarkably, SFPQ-TFE3 expression is sufficient to induce lineage plasticity in renal tubular epithelial cells, with rapid down-regulation of the critical PAX2/PAX8 nephric lineage factors and tubular epithelial markers, and concomitant up-regulation of PEComa differentiation markers in transgenic mice, human cell line models and human tRCC. Pharmacologic or genetic inhibition of mTOR signaling downregulates expression of the SFPQ-TFE3 fusion protein and rescues nephric lineage marker expression and transcriptional activity in vitro. These data provide evidence of a potential epithelial cell-of-origin for TFE3 -driven PEComas and highlight a reciprocal role for SFPQ-TFE3 and mTOR in driving lineage plasticity in the kidney, expanding our understanding of the pathogenesis of MiT/TFE-driven tumors.
Collapse
|
2
|
Qu Y, Wu X, Anwaier A, Feng J, Xu W, Pei X, Zhu Y, Liu Y, Bai L, Yang G, Tian X, Su J, Shi GH, Cao DL, Xu F, Wang Y, Gan HL, Ni S, Sun MH, Zhao JY, Zhang H, Ye D, Ding C. Proteogenomic characterization of MiT family translocation renal cell carcinoma. Nat Commun 2022; 13:7494. [PMID: 36470859 PMCID: PMC9722939 DOI: 10.1038/s41467-022-34460-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/26/2022] [Indexed: 12/12/2022] Open
Abstract
Microphthalmia transcription factor (MiT) family translocation renal cell carcinoma (tRCC) is a rare type of kidney cancer, which is not well characterized. Here we show the comprehensive proteogenomic analysis of tRCC tumors and normal adjacent tissues to elucidate the molecular landscape of this disease. Our study reveals that defective DNA repair plays an important role in tRCC carcinogenesis and progression. Metabolic processes are markedly dysregulated at both the mRNA and protein levels. Proteomic and phosphoproteome data identify mTOR signaling pathway as a potential therapeutic target. Moreover, molecular subtyping and immune infiltration analysis characterize the inter-tumoral heterogeneity of tRCC. Multi-omic integration reveals the dysregulation of cellular processes affected by genomic alterations, including oxidative phosphorylation, autophagy, transcription factor activity, and proteasome function. This study represents a comprehensive proteogenomic analysis of tRCC, providing valuable insights into its biological mechanisms, disease diagnosis, and prognostication.
Collapse
Affiliation(s)
- Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Xiaohui Wu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Aihetaimujiang Anwaier
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Jinwen Feng
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Xiaoru Pei
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Yang Liu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Lin Bai
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Guojian Yang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Jiaqi Su
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Guo-Hai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Da-Long Cao
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Fujiang Xu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yue Wang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
| | - Hua-Lei Gan
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Meng-Hong Sun
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China
- Tissue Bank & Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
- Department of Oncology, Shanghai Medical College, Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
| | - Chen Ding
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Sun H, Wei X, Zeng C. Autophagy in Xp11 translocation renal cell carcinoma: from bench to bedside. Mol Cell Biochem 2021; 476:4231-4244. [PMID: 34345999 DOI: 10.1007/s11010-021-04235-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/27/2021] [Indexed: 12/25/2022]
Abstract
Xp11 translocation renal cell carcinoma (tRCC) characterized by the rearrangement of the TFE3 is recently identified as a unique subtype of RCC that urgently requires effective prevention and treatment strategies. Therefore, determining suitable therapeutic targets and fully understanding the biological significance of tRCC is essential. The importance of autophagy is increasingly acknowledged because it shows carcinogenic activity or suppressor effect. Autophagy is a physiological cellular process critical to maintaining cell homeostasis, which is involved in the lysosomal degradation of cytoplasmic organelles and macromolecules via the lysosomal pathway, suggesting that targeting autophagy is a potential therapeutic approach for cancer therapies. However, the underlying mechanism of autophagy in tRCC is still ambiguous. In this review, we summarize the autophagy-related signaling pathways associated with tRCC. Moreover, we examine the roles of autophagy and the immune response in tumorigenesis and investigate how these factors interact to facilitate or prevent tumorigenesis. Besides, we review the findings regarding the treatment of tRCC via induction or inhibition of autophagy. Hopefully, this study will shed some light on the functions and implications of autophagy and emphasize its role as a potential molecular target for therapeutic intervention in tRCC.
Collapse
Affiliation(s)
- Huimin Sun
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
| | - Xing Wei
- Department of Nephrology and Rheumatology, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, 518110, China.
| |
Collapse
|
4
|
Singh AC, Pal M, Kapoor A, Menon N, Prabhash K, Noronha V, Bakshi G, Prakash G, Menon S, Sable N, Kalra D, Kulkarni S, Joshi A. Study of Treatment Outcome in Adults with TFE-Related RCC. South Asian J Cancer 2021; 10:92-96. [PMID: 34568222 PMCID: PMC8460340 DOI: 10.1055/s-0041-1731264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Introduction
TFE Translocation renal cell carcinoma (TRCC) represents 1 to 5% of all cases of renal cell carcinoma, with the highest frequency among children and young adults. Management of these tumors is not very well defined in literature. Although in pediatric age group it has favorable prognosis, in adults it has an aggressive nature, with poor outcome. This is a retrospective analysis of treatment outcome in adult patient 18 years or above treated at our hospital between January 2013 and November 2018.
Material and Methods
Clinical and pathological data of 26 patients from a single institution diagnosed with TRCC between January 2013 and November 2018 were retrospectively reviewed. All cases of TRCC were confirmed with immunohistochemistry or fluorescence in situ hybridization. We analyzed our data of patients treated with surgery only or who progressed after surgery and treated with systemic therapy or who presented with upfront unresectable or metastatic disease treated with systemic therapy with respect to event-free survival (EFS) and overall survival (OS).
Results
Between January 2013 and November 2018, 26 adult patients who were treated at our center were eligible for this analysis as per our criteria. Out of 26 patients, 25 patients had radical surgery after evaluation and 1 had metastatic disease who was started on systemic therapy. Out 25 patients who were treated with radical surgery, 16 patients progressed and they were started on systemic therapy except for 1 patient who defaulted. Median time to start systemic therapy among patient treated with curative nephrectomy was 13 months. Median EFS and median OS among overall population were 22 and 30 months, respectively. Among 16 patients who were treated with systemic therapy, median EFS to first-line therapy was 8 months and to second-line therapy was 2.5 months. Median OS was 17 months in patients treated with systemic therapy.
Conclusion
TRCC is rare in adult population but carries significant risk of disease progression even after initial curative treatment with potential response to targeted therapy for short duration.
Collapse
Affiliation(s)
- Ajaykumar Chandrabhan Singh
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mahendra Pal
- Department of Urologic Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akhil Kapoor
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Center and Homi Bhabha Cancer Hospital (A Unit of Tata Memorial Center, Mumbai), Varanasi, Uttar Pradesh, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ganesh Bakshi
- Department of Urologic Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gagan Prakash
- Department of Urologic Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Santosh Menon
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nilesh Sable
- Department of Radiology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Devanshi Kalra
- CRS, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sheetal Kulkarni
- CRS, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
5
|
Wu Y, Chen S, Zhang M, Liu K, Jing J, Pan K, Zhang L, Xu B, Lu X, Chen M. Factors Associated with Survival From Xp11.2 Translocation Renal Cell Carcinoma Diagnosis-A Systematic Review and Pooled Analysis. Pathol Oncol Res 2021; 27:610360. [PMID: 34257577 PMCID: PMC8262176 DOI: 10.3389/pore.2021.610360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/03/2021] [Indexed: 12/15/2022]
Abstract
Purpose: Xp11.2 translocation renal cell carcinoma (Xp11.2 tRCC) is a rare subtype of renal cell carcinoma (RCC), characterized by translocations of Xp11.2 breakpoints, involving of the transcription factor three gene (TFE3). The aim of our study was to comprehensively characterize the clinical characteristics and outcomes, and to identify risk factors associated with OS and PFS in Xp11.2 tRCC patients. Methods: Literature search on Xp11.2 tRCC was performed using databases such as pubmed EMBASE and Web of Science. Studies were eligible if outcomes data (OS and/or PFS) were reported for patients with a histopathologically confirmed Xp11.2 tRCC. PFS and OS were evaluated using the univariable and multivariable Cox regression model. Results: There were 80 eligible publications, contributing 415 patients. In multivariable analyses, the T stage at presentation was significantly associated with PFS (HR: 3.87; 95% CI: 1.70 to 8.84; p = 0.001). The median time of PFS was 72 months. In the multivariable analyses, age at diagnosis (HR: 2.16; 95% CI: 1.03 to 4.50; p = 0.041), T stage at presentation (HR: 4.44; 95% CI: 2.16 to 9.09; p < 0.001) and metastasis status at presentation (HR: 2.67; 95% CI: 1.12 to 6.41; p = 0.027) were all associated with OS, with a median follow-up time of 198 months. Conclusion: T stage at presentation is the only factor that is associated with both PFS and OS in patients with Xp11.2 tRCC. Also, patients over 45 or with metastases are more likely to have poorer OS.
Collapse
Affiliation(s)
- Yuqing Wu
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Saisai Chen
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Minhao Zhang
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Kuangzheng Liu
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Jibo Jing
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Kehao Pan
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Lihua Zhang
- Department of Pathology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Bin Xu
- Surgical Research Center, Institute of Urology, School of Medicine, Southeast University, Nanjing, China.,Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China
| | - Xiaoming Lu
- Department of Urology, Yancheng Third People's Hospital, Yancheng, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, China.,Lishui People's Hospital, Nanjing, China
| |
Collapse
|
6
|
Geller JI, Cost NG, Chi YY, Tornwall B, Cajaiba M, Perlman EJ, Kim Y, Mullen EA, Glick RD, Khanna G, Daw NC, Ehrlich P, Fernandez CV, Dome JS. A prospective study of pediatric and adolescent renal cell carcinoma: A report from the Children's Oncology Group AREN0321 study. Cancer 2020; 126:5156-5164. [PMID: 32926409 DOI: 10.1002/cncr.33173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/06/2020] [Accepted: 07/17/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND To the authors' knowledge, AREN0321 is the first prospective clinical study of pediatric and adolescent renal cell carcinoma (RCC). Goals of the study included establishing epidemiological, treatment, and outcome data and confirming that patients with completely resected pediatric RCC, including lymph node-positive disease (N1), have a favorable prognosis without adjuvant therapy. METHODS From 2006 to 2012, patients aged <30 years with centrally reviewed pathology of RCC were enrolled prospectively. RESULTS A total of 68 patients were enrolled (39 of whom were male; median age of 13 years [range, 0.17-22.1 years]). Stage was classified according to the American Joint Committee on Cancer TNM stage seventh edition as stage I in 26 patients, stage II in 7 patients, stage III in 26 patients, and stage IV in 8 patients, and was not available in 1 patient. Sixty patients underwent resection of all known sites of disease, including 2 patients with stage IV disease. Surgery included radical nephrectomy (53 patients [81.5%]), partial nephrectomy (12 patients [18.5%]), and unknown (3 patients [4.4%]). Histology was TFE-associated RCC (translocation-type RCC; tRCC) in 40 patients, RCC not otherwise specified and/or other in 13 patients, papillary RCC in 9 patients, and renal medullary carcinoma (RMC) in 6 patients. Lymph node status was N0 in 21 patients, N1 in 21 patients (tRCC in 15 patients, RMC in 3 patients, papillary RCC in 2 patients, and not otherwise specified and/or other in 1 patient), and Nx in 26 patients. The 4-year event-free survival and overall survival rates were 80.2% (95% CI, 69.6%-90.9%) and 84.8% (95% CI, 75.2%-94.5%), respectively, overall and 87.5% (95% CI, 68.3%-100%) and 87.1% (95% CI, 67.6%-100%), respectively, for the 16 patients with N1M0 disease. Among patients presenting with metastases, 2 of 8 patients (2 of 5 patients with RMC) were alive (1 with disease) at the time of last follow-up, including 1 patient who was lost to follow-up (succinate dehydrogenase deficiency). The predominant RCC subtypes associated with mortality were tRCC and RMC. CONCLUSIONS Favorable short-term outcomes can be achieved without adjuvant therapy in children and adolescents with completely resected RCC, independent of lymph node status. A prospective study of patients with tRCC and RMC with M1 or recurrent disease is needed to optimize treatment.
Collapse
Affiliation(s)
- James I Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine, the Children's Hospital Colorado, Aurora, Colorado
| | - Yueh-Yun Chi
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, Florida
| | - Brett Tornwall
- Department of Biostatistics, Children's Oncology Group Statistics and Data Center, University of Florida, Gainesville, Florida
| | - Mariana Cajaiba
- Department of Pathology, Anne and Robert H. Lurie Children's Hospital, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Elizabeth J Perlman
- Department of Pathology, Ann and Robert H. Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yeonil Kim
- Biostatistics and Research Decision Sciences, Merck Research Laboratories, Merck & Company Inc, Rahway, New Jersey
| | - Elizabeth A Mullen
- Department of Pediatric Oncology, Dana-Farber Cancer Institute/Boston Children's Hospital, Boston, Massachusetts
| | - Richard D Glick
- Division of Pediatric Surgery, Steven and Alexandra Cohen Medical Center of New York, New York, New York
| | - Geetika Khanna
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | - Najat C Daw
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peter Ehrlich
- Section of Pediatric Surgery, Department of Surgery, C.S. Mott Children's Hospital, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Conrad V Fernandez
- Division of Pediatric Oncology, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Jeffrey S Dome
- Division of Pediatric Oncology, Children's National Hospital, George Washington University School of Medicine and Health Sciences, Washington, DC
| | | |
Collapse
|
7
|
Abstract
Pediatric and adolescent renal tumors account for approximately 7% of all new cancer diagnoses in the USA each year. The prognosis and treatment are varied based on factors including the underlying histology and tumor stage, with survival rates ranging from greater than 90% in favorable histology Wilms tumor to almost universally fatal in other disease types, including those patients with advanced stage malignant rhabdoid tumor and renal medullary carcinoma. In recent years, our understanding of the underlying genetic drivers of the different types of pediatric kidney cancer has dramatically increased, opening the door to utilization of new targeted biologic agents alone or in combination with conventional chemotherapy to improve outcomes. Several ongoing clinical trials are investigating the use of a variety of targeted agents in pediatric patients with underlying genetic aberrations. In this manuscript, the underlying biology and early phase clinical trials relevant to pediatric renal cancers are reviewed.
Collapse
Affiliation(s)
- Amy L Walz
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA.
| | | | - James I Geller
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
8
|
Kauffman EC, Lang M, Rais-Bahrami S, Gupta GN, Wei D, Yang Y, Sourbier C, Srinivasan R. Preclinical efficacy of dual mTORC1/2 inhibitor AZD8055 in renal cell carcinoma harboring a TFE3 gene fusion. BMC Cancer 2019; 19:917. [PMID: 31519159 PMCID: PMC6743205 DOI: 10.1186/s12885-019-6096-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022] Open
Abstract
Background Renal cell carcinomas (RCC) harboring a TFE3 gene fusion (TfRCC) represent an aggressive subset of kidney tumors. Key signaling pathways of TfRCC are unknown and preclinical in vivo data are lacking. We investigated Akt/mTOR pathway activation and the preclinical efficacy of dual mTORC1/2 versus selective mTORC1 inhibition in TfRCC. Methods Levels of phosphorylated Akt/mTOR pathway proteins were compared by immunoblot in TfRCC and clear cell RCC (ccRCC) cell lines. Effects of the mTORC1 inhibitor, sirolimus, and the dual mTORC1/2 inhibitor, AZD8055, on Akt/mTOR activation, cell cycle progression, cell viability and cytotoxicity were compared in TfRCC cells. TfRCC xenograft tumor growth in mice was evaluated after 3-week treatment with oral AZD8055, intraperitoneal sirolimus and respective vehicle controls. Results The Akt/mTOR pathway was activated to a similar or greater degree in TfRCC than ccRCC cell lines and persisted partly during growth factor starvation, suggesting constitutive activation. Dual mTORC1/2 inhibition with AZD8055 potently inhibited TfRCC viability (IC50 = 20-50 nM) due at least in part to cell cycle arrest, while benign renal epithelial cells were relatively resistant (IC50 = 400 nM). Maximal viability reduction was greater with AZD8055 than sirolimus (80–90% versus 30–50%), as was the extent of Akt/mTOR pathway inhibition, based on significantly greater suppression of P-Akt (Ser473), P-4EBP1, P-mTOR and HIF1α. In mouse xenograft models, AZD8055 achieved significantly better tumor growth inhibition and prolonged mouse survival compared to sirolimus or vehicle controls. Conclusions Akt/mTOR activation is common in TfRCC and a promising therapeutic target. Dual mTORC1/2 inhibition suppresses Akt/mTOR signaling more effectively than selective mTORC1 inhibition and demonstrates in vivo preclinical efficacy against TFE3-fusion renal cell carcinoma. Electronic supplementary material The online version of this article (10.1186/s12885-019-6096-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eric C Kauffman
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Departments of Urology and Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY, 14263, USA
| | - Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Soroush Rais-Bahrami
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Department of Urology and Department of Radiology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35294, USA
| | - Gopal N Gupta
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Department of Urology, Loyola University Medical Center, Chicago, IL, 60153, USA
| | - Darmood Wei
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Youfeng Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.,Present address: Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 10 - Hatfield CRC, Room 1-5940, Bethesda, MD, 20892, USA.
| |
Collapse
|
9
|
Ambalavanan M, Geller JI. Treatment of advanced pediatric renal cell carcinoma. Pediatr Blood Cancer 2019; 66:e27766. [PMID: 31012542 DOI: 10.1002/pbc.27766] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 04/04/2019] [Accepted: 04/05/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Pediatric renal cell carcinoma (pRCC) is the second most common renal malignancy of childhood; however, treatment data for advanced disease is lacking. METHODS A retrospective analysis of pRCC patients (age < 21 years at diagnosis) treated between 2000 and 2015 at Cincinnati Children's Hospital Medical Center was undertaken, with specific focus on medical therapies, accompanied by a detailed literature review. RESULTS Twenty-four patients (median age = 15 years) were identified; 11 were female. Past history of kidney pathology (4) and prior hematologic/oncologic diagnoses (5) were common associated findings. Translocation morphology RCC (tRCC) was the most common subtype (16; 64%), followed by papillary (6; 24%), clear cell renal cell carcinoma (ccRCC) (1), and chromophobe (1). The TNM stage distribution was I (8; 33%), II (2; 8%), III (3; 13%), and IV (11; 46%). Eleven patients with stage IV disease all had tRCC and received medicinal anticancer therapies, the most common being antiangiogenic (10), conventional chemotherapy (8), mTOR inhibition (7), and immunotherapy (3). Four patients also received small-port radiotherapy. The mean time to progression (TTP) was longest for axitinib (n = 2; TTP = 7.8 m; range 5.5-10 m) and sunitinib (n = 6; TTP = 4.7 m; range 0.3-12 m). Overall, 20 cases of pediatric RCC who received RCC-directed medicinal therapy with outcome data have been previously reported. CONCLUSIONS For patients with unresectable pRCC requiring systemic therapy, available data are scarce. Data herein support an increased TTP with antiangiogenic therapy in tRCC supporting a formal study of antiangiogenic therapies through multicooperative-group collaboration.
Collapse
Affiliation(s)
| | - James I Geller
- Division of Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
10
|
Rua Fernández OR, Escala Cornejo R, Navarro Martín M, García Muñoz M, Antunez Plaza P, García Dominguez AR, Cruz Hernández JJ. Renal Cell Carcinoma Associated With Xp11.2 Translocation/TFE3 Gene-fusion: A Long Response to mammalian target of rapamycin (mTOR) Inhibitors. Urology 2018; 117:41-43. [PMID: 29702156 DOI: 10.1016/j.urology.2018.03.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/04/2018] [Accepted: 03/22/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To demonstrate that patients with Xp11.2/TFE3 gene-fusion translocation renal cell carcinoma (RCC), despite having an aggressive course in young adults, could have valid treatment options such as mammalian target of rapamycin (mTOR) inhibitors with good outcomes. Furthermore, to explain possible mechanisms of action of mTOR inhibitors in this type of RCC. MATERIALS AND METHODS We report a case of a 44-year-old man who has been treated with everolimus for a Xp11.2 translocation/TFE3 gene-fusion RCC after 2 previous failed treatments with tyrosine kinase inhibitor. During the follow-up, we evaluated type and duration of response with everolimus. RESULTS The patient obtained a long-lasting response of disease of 25 months with everolimus without any symptom. CONCLUSION We believe that mTOR inhibitors could be a good line option treatment to consider for this type of patients.
Collapse
Affiliation(s)
- Oliver R Rua Fernández
- Department of Medical Oncology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Roberto Escala Cornejo
- Department of Medical Oncology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain.
| | - Miguel Navarro Martín
- Department of Medical Oncology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - María García Muñoz
- Department of Medical Oncology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | - Patricia Antunez Plaza
- Department of Pathology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | | | - Juan J Cruz Hernández
- Department of Medical Oncology, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| |
Collapse
|
11
|
Verma SP, Agarwal A, Das P. Sodium butyrate induces cell death by autophagy and reactivates a tumor suppressor gene DIRAS1 in renal cell carcinoma cell line UOK146. In Vitro Cell Dev Biol Anim 2018; 54:295-303. [PMID: 29556894 DOI: 10.1007/s11626-018-0239-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/21/2018] [Indexed: 12/01/2022]
Abstract
Sodium butyrate (SB), a histone deacetylase inhibitor, is emerging as a potent anti-cancer drug for different types of cancers. In the present study, anti-cancer activity of SB in Xp11.2 (TFE3) translocated renal cell carcinoma cell line UOK146 was studied. Anti-proliferative effect of SB in renal cell carcinoma (RCC) cell line UOK146 was evaluated by MTT assay and morphological characteristics were observed by phase contrast microscopy which displayed the cell death after SB treatment. SB induces DNA fragmentation and change in nuclear morphology observed by increased sub-G1 region cell population and nuclear blebbings. Cell cycle arrest at G2/M phase was found after SB treatment. UOK146 cell line shows autophagy mode of cell death as displayed by acridine orange staining and flow cytometry analysis. LC3-II, a protein marker of autophagy, was also found to be upregulated after SB treatment. A tumor suppressor gene DIRAS1 was upregulated after SB treatment, displaying its anti-cancer potential at molecular level. These findings suggest that SB could serve as a novel regulator of tumor suppressors and lead to the discovery of novel therapeutics with better and enhanced anti-cancer activity.
Collapse
Affiliation(s)
- Shiv Prakash Verma
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | | | - Parimal Das
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
12
|
Fang Y, Bao W, Rao Q, Wang X, Xia Q, Shen Q, Zhou X, Yao B. TFE3 regulates renal adenocarcinoma cell proliferation via activation of the mTOR pathway. Mol Med Rep 2017; 16:2721-2725. [PMID: 28713889 PMCID: PMC5547915 DOI: 10.3892/mmr.2017.6930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/15/2017] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to investigate the role of transcription factor E3 (TFE3) in the regulation of proliferation in renal adenocarcinoma cells. The LV-TFE3 overexpression (OE) lentivirus and negative control CON195 (NC) lentivirus were transfected into the ACHN cell line. Protein expression of FLAG-tag TFE3 was determined using western blot analysis. Differences in cell proliferation, plate clone formation and cell cycle distribution between OE and NC groups were compared using MTT, plate colony formation and flow cytometry assays, respectively. The levels of mammalian target of rapamycin (mTOR) and phosphorylated ribosomal protein S6 (p-rpS6) were analyzed by western blotting. Cell proliferation and colony formation increased significantly in the OE group compared with the NC group. The % of cells in the G1 and G2 phases of the cell cycle decreased, while the % of cells in the S phase of the cell cycle increased in the OE group compared with the NC group. In addition, mTOR and p-rpS6 levels were increased in the OE group compared with the NC group. The results of the present study demonstrated that TFE3 overexpression resulted in increased ACHN cell proliferation and plate clone formation. TFE3 may promote renal tumor growth by regulating cell cycle progression and activating the phosphatidylinositol 3-kinase/AKT serine/threonine kinase 1/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yuan Fang
- Reproductive Medical Center, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Bao
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Xuan Wang
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiuyuan Xia
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Qin Shen
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiaojun Zhou
- Department of Pathology, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| | - Bing Yao
- Reproductive Medical Center, Jinling Hospital, Clinical Medical School of Southern Medical University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
13
|
Kakoki K, Miyata Y, Mochizuki Y, Iwata T, Obatake M, Abe K, Nagayasu T, Sakai H. Long-term Treatment With Sequential Molecular Targeted Therapy for Xp11.2 Translocation Renal Cell Carcinoma: A Case Report and Review of the Literature. Clin Genitourin Cancer 2017; 15:e503-e506. [DOI: 10.1016/j.clgc.2016.12.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/14/2016] [Accepted: 12/22/2016] [Indexed: 12/22/2022]
|
14
|
Calcagnì A, Kors L, Verschuren E, De Cegli R, Zampelli N, Nusco E, Confalonieri S, Bertalot G, Pece S, Settembre C, Malouf GG, Leemans JC, de Heer E, Salvatore M, Peters DJ, Di Fiore PP, Ballabio A. Modelling TFE renal cell carcinoma in mice reveals a critical role of WNT signaling. eLife 2016; 5. [PMID: 27668431 PMCID: PMC5036965 DOI: 10.7554/elife.17047] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 08/15/2016] [Indexed: 12/16/2022] Open
Abstract
TFE-fusion renal cell carcinomas (TFE-fusion RCCs) are caused by chromosomal translocations that lead to overexpression of the TFEB and TFE3 genes (Kauffman et al., 2014). The mechanisms leading to kidney tumor development remain uncharacterized and effective therapies are yet to be identified. Hence, the need to model these diseases in an experimental animal system (Kauffman et al., 2014). Here, we show that kidney-specific TFEB overexpression in transgenic mice, resulted in renal clear cells, multi-layered basement membranes, severe cystic pathology, and ultimately papillary carcinomas with hepatic metastases. These features closely recapitulate those observed in both TFEB- and TFE3-mediated human kidney tumors. Analysis of kidney samples revealed transcriptional induction and enhanced signaling of the WNT β-catenin pathway. WNT signaling inhibitors normalized the proliferation rate of primary kidney cells and significantly rescued the disease phenotype in vivo. These data shed new light on the mechanisms underlying TFE-fusion RCCs and suggest a possible therapeutic strategy based on the inhibition of the WNT pathway.
Collapse
Affiliation(s)
- Alessia Calcagnì
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Lotte Kors
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Pathology, Academical Medical Center, Amsterdam, The Netherlands
| | - Eric Verschuren
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Nicolina Zampelli
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Edoardo Nusco
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy
| | - Stefano Confalonieri
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, Milan, Italy
| | - Giovanni Bertalot
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy
| | - Salvatore Pece
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Settembre
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, United States.,Medical Genetics, Federico II University, Naples, Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy
| | - Gabriel G Malouf
- Department of Medical Oncology Groupe Hospitalier Pitie-Salpetriere, University Paris 6, Paris, France.,Assistance Publique Hopitaux de Paris, University Paris 6, Paris, France.,Faculty of Medicine Pierre et Marie Curie, University Paris 6, Paris, France.,Institut Universitaire de Cancerologie GRC5, University Paris 6, Paris, France
| | - Jaklien C Leemans
- Department of Pathology, Academical Medical Center, Amsterdam, The Netherlands
| | - Emile de Heer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Dorien Jm Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Pier Paolo Di Fiore
- Molecular Medicine Program, European Institute of Oncology, Milan, Italy.,IFOM, The FIRC Institute for Molecular Oncology Foundation, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine, TIGEM, Pozzuoli, Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, United States.,Medical Genetics, Federico II University, Naples, Italy.,Medical Genetics, Department of Medical and Translational Sciences, Federico II University, Naples, Italy
| |
Collapse
|
15
|
Chen X, Zhu Q, Li B, Cui W, Zhou H, Duan N, Liu Y, Kundra V, Wang Z. Renal cell carcinoma associated with Xp11.2 translocation/TFE gene fusion: imaging findings in 21 patients. Eur Radiol 2016; 27:543-552. [DOI: 10.1007/s00330-016-4421-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 02/29/2016] [Accepted: 05/19/2016] [Indexed: 02/03/2023]
|
16
|
Noh BJ, Kim YW, Chang SG, Park YK. A rare Xp11 translocation carcinoma with ossification: a case report with morphoproteomic insights into bone formation. Pathology 2016; 48:383-6. [PMID: 27131933 DOI: 10.1016/j.pathol.2016.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 02/19/2016] [Accepted: 03/01/2016] [Indexed: 11/25/2022]
Affiliation(s)
- Byeong-Joo Noh
- Department of Pathology, Kyung Hee University Medical Center, Korea
| | - Youn Wha Kim
- Department of Pathology, School of Medicine, Kyung Hee University, Korea
| | - Sung-Goo Chang
- Department of Urology, Kyung Hee University Medical Center, Seoul, Korea
| | - Yong-Koo Park
- Department of Pathology, School of Medicine, Kyung Hee University, Korea.
| |
Collapse
|
17
|
Phosphorylated 4EBP1 is associated with tumor progression and poor prognosis in Xp11.2 translocation renal cell carcinoma. Sci Rep 2016; 6:23594. [PMID: 27026382 PMCID: PMC4812307 DOI: 10.1038/srep23594] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/09/2016] [Indexed: 12/27/2022] Open
Abstract
Two main signaling pathways, PI3K-AKT-mTOR and RAS-MAPK, are involved in transmitting the proliferative signals which play critical roles in human cancers. However, the functions of these pathways in Xp11.2 RCC remain undefined. This study aimed to explore the expression of mTOR and MAPK cascades in Xp11.2 RCC and to assess the prognostic significance of proteins evaluated. Immunohistochemistry was performed to evaluate the expression of 4EBP1, p-4EBP1, p-mTOR, p-S6K and p-MAPK in 36 adult Xp11.2 RCC patients who were confirmed by FISH assay. Cox regression models were used to evaluate the prognostic value of all covariates. Among 36 assessed patients, 14 (38.9%), 26 (72.2%), 16 (44.4%), 19 (52.8%), and 9 (25.0%) patients showed high expression of 4EBP1, p-4EBP1, p-mTOR, p-S6K, and p-MAPK, respectively. We noted that p-4EBP1 expression was significantly correlated with lymph node metastases (P = 0.027). Multivariate analysis showed that high p-4EBP1 expression was an independent adverse prognostic factor for both PFS (HR = 33.750, P = 0.017) and OS (HR = 56.111, P = 0.026). Our findings suggest that p-4EBP1 may serve as a funnel factor that converge the upstream proliferative oncogenic signals. Effective inhibition of the pathways responsible for 4E-BP1 phosphorylation might be a useful strategy to improve the outcome of Xp11.2 RCC patients.
Collapse
|
18
|
Geller JI, Ehrlich PF, Cost NG, Khanna G, Mullen EA, Gratias EJ, Naranjo A, Dome JS, Perlman EJ. Characterization of adolescent and pediatric renal cell carcinoma: A report from the Children's Oncology Group study AREN03B2. Cancer 2015; 121:2457-64. [PMID: 25845370 DOI: 10.1002/cncr.29368] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 11/08/2022]
Abstract
BACKGROUND The current study was conducted to characterize the epidemiology, histology, and radiographic features of as well as the surgical approach to pediatric and adolescent renal cell carcinoma (pRCC). METHODS pRCC cases prospectively enrolled on the Children's Oncology Group study AREN03B2 underwent central pathology, radiology, surgery, and oncology review. RESULTS As of June 2012, 120 of a total of 3250 patients enrolled on AREN03B2 (3.7%) were found to have unilateral RCC (median age, 12.9 years [range, 1.9-22.1 years]; 52.5% were female). Central review classified these as translocation morphology (56 patients), papillary (20 patients), renal medullary carcinoma (13 patients), chromophobe (4 patients), oncocytoma (1 patient), conventional clear cell (1 patient), and RCC not otherwise specified (25 patients). Lymph node (LN) involvement (N+) was found in 35 of 73 cases (47.9%) for which LNs were sampled, including 19 of 40 cases with primary tumors measuring <7 cm (47.5%). Using a size cutoff of 1 cm, imaging detection of LN involvement had a sensitivity of 57.14% (20 of 35 cases; 95% CI, 39.35%-73.68%) and a specificity of 94.59% (35 of 37 cases; 95% CI, 81.81%-99.34%). Distant metastases were present in 23 cases (19.2%). Initial surgery was radical nephrectomy in 88 patients (73.3%), nephron-sparing surgery in 18 patients (15.0%), and biopsy in 14 patients (11.7%). Compared with patients undergoing radical nephrectomy, those treated with nephron-sparing surgery were less likely to have LNs sampled (6 of 18 patients [33.3%] vs 65 of 88 patients [73.9%]; P = .002). CONCLUSIONS Translocation RCC is the most common form of pediatric and adolescent RCC. Lymph node disease is common and observed among patients with small primary tumors. Imaging has a high specificity but relatively low sensitivity for the detection of such lymph node disease. Failure to sample LNs results in incomplete staging and potentially inadequate disease control for younger patients with RCC.
Collapse
Affiliation(s)
- James I Geller
- Division of Pediatric Oncology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Peter F Ehrlich
- Section of Pediatric Surgery, Department of Surgery, CS Mott Children's Hospital, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Nicholas G Cost
- Division of Urology, Department of Surgery, University of Colorado School of Medicine and the Children's Hospital Colorado, Aurora, Colorado
| | - Geetika Khanna
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Elizabeth A Mullen
- Department of Pediatric Oncology, Children's Hospital Boston/Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eric J Gratias
- Division of Pediatric Hematology/Oncology, T.C. Thompson Children's Hospital, University of Tennessee College of Medicine-Chattanooga, Chattanooga, Tennessee
| | - Arlene Naranjo
- Department of Biostatistics, University of Florida, Children's Oncology Group Statistics and Data Center, Gainesville, Florida
| | - Jeffrey S Dome
- Division of Pediatric Oncology, Children's National Medical Center, Washington, DC
| | - Elizabeth J Perlman
- Department of Pathology, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
19
|
Su HH, Sung MT, Chiang PH, Cheng YT, Chen YT. The preliminary experiences of translocation renal cell carcinoma and literature review. Kaohsiung J Med Sci 2014; 30:402-8. [PMID: 25002378 DOI: 10.1016/j.kjms.2014.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 12/13/2013] [Accepted: 01/07/2014] [Indexed: 11/24/2022] Open
Abstract
Xp11.2 translocation renal cell carcinoma (RCC) is rare and predominantly found in children and young adults. Because of the property of overexpressed transcription factor E3 (TFE3) fusion protein, immunohistochemical (IHC) staining with TFE3 antibody makes an excellent diagnostic tool. This study analyzed preliminary experiences of eight Xp11.2 translocation RCCs in our institution between 2007 and 2012. In four males and four females with a mean age of 28.4 years. Xp11.2 translocation RCCs were diagnosed. TFE3 IHC stain was positive in all tumor specimens. As the initial presentation, four patients suffered from abdominal pain, three cases had gross hematuria, and one case had hemoptysis caused by existing lung metastasis. The tumor was located in the right kidney (75%) with mean diameter of 5.85 ± 2.64 cm. Three cases (38%, 3/8) presented with lymph node metastasis at the time of diagnosis. In five cases (63%, 5/8), the initial diagnosis was Stage III and IV. Treatment included open surgery (one partial nephrectomy and five radical nephrectomies), cryoablation, immunotherapy, and target therapy. The mean follow-up time was 32 months. One patient died after 23.4 months of follow-up. The application of TFE3 IHC stain will improve the diagnostic accuracy for detecting XP11.2 translocation renal cell carcinoma. Surgery or cryoablation both had excellent prognosis in early stages. Although the disease is believed to be indolent, an increasingly aggressive clinical course should be kept in mind, especially for children and young adults.
Collapse
Affiliation(s)
- Hsin-Hao Su
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ming-Tse Sung
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Po-Hui Chiang
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Yunan-Tso Cheng
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yen-Ta Chen
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Dome JS, Fernandez CV, Mullen EA, Kalapurakal JA, Geller JI, Huff V, Gratias EJ, Dix DB, Ehrlich PF, Khanna G, Anderson JR, Naranjo A, Perlman EJ, Perlman EJ. Children's Oncology Group's 2013 blueprint for research: renal tumors. Pediatr Blood Cancer 2013; 60:994-1000. [PMID: 23255438 PMCID: PMC4127041 DOI: 10.1002/pbc.24419] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 11/07/2012] [Indexed: 12/12/2022]
Abstract
Renal malignancies are among the most prevalent pediatric cancers. The most common is favorable histology Wilms tumor (FHWT), which has 5-year overall survival exceeding 90%. Other pediatric renal malignancies, including anaplastic Wilms tumor, clear cell sarcoma, malignant rhabdoid tumor, and renal cell carcinoma, have less favorable outcomes. Recent clinical trials have identified gain of chromosome 1q as a prognostic marker for FHWT. Upcoming studies will evaluate therapy adjustments based on this and other novel biomarkers. For high-risk renal tumors, new treatment regimens will incorporate biological therapies. A research blueprint, viewed from the perspective of the Children's Oncology Group, is presented.
Collapse
Affiliation(s)
- Jeffrey S. Dome
- Division of Oncology, Center for Cancer and Blood Disorders, Children’s National Medical Center, Washington, DC,Correspondence to: Jeffrey S. Dome, MD, PhD, Division of Oncology, Center for Cancer and Blood Disorders, Children’s National Medical Center, 111 Michigan Avenue NW, Washington DC 20010,
| | | | | | - John A. Kalapurakal
- Department of Radiation Oncology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL
| | - James I. Geller
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Vicki Huff
- Department of Genetics, MD Anderson Cancer Center, Houston, TX
| | - Eric J. Gratias
- Division of Hematology/Oncology, Children’s Hospital at Erlanger, University of Tennessee College of Medicine, Chattanooga, TN
| | - David B. Dix
- Pediatric Hematology/Oncology, British Columbia Children’s Hospital, Vancouver, BC
| | - Peter F. Ehrlich
- Department of Pediatric Surgery, University of Michigan, CS Mott Children’s Hospital, Ann Arbor, MI
| | - Geetika Khanna
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO
| | | | - Arlene Naranjo
- Department of Biostatistics, Colleges of Medicine and Public Health & Health Professions, University of Florida, Gainesville, Florida
| | - Elizabeth J. Perlman
- Department of Pathology, Northwestern University’s Feinberg School of Medicine and the Robert H. Lurie Cancer Center, Chicago, IL
| | | | | |
Collapse
|
21
|
Zhu QQ, Wang ZQ, Zhu WR, Chen WX, Wu JT. The multislice CT findings of renal carcinoma associated with XP11.2 translocation/TFE gene fusion and collecting duct carcinoma. Acta Radiol 2013; 54:355-62. [PMID: 23446748 DOI: 10.1258/ar.2012.120255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Renal cell carcinoma associated with Xp11.2 translocation and TFE gene fusion (Xp11.2/TFE RCC), and collecting duct carcinoma (CDC) are uncommon subtypes of renal cell carcinomas. PURPOSE To investigate the multislice CT (MSCT) characteristics of these two tumor types. MATERIAL AND METHODS Nine patients with Xp11.2/TFE RCC and 10 patients with CDC were studied retrospectively. MSCT was undertaken to investigate differences in tumor characteristics and enhancement patterns. RESULTS All patients had single tumors centered in the renal medulla. Two patients with each tumor type had lymph node involvement and there was a single case of hepatic metastasis (Xp11.2/TFE RCC). The mean tumor diameter of Xp11.2/TFE RCC tumors was significantly larger than for CDC tumors. Two patients with Xp11.2/TFE RCC had cystic components as did eight patients with CDC (P < 0.05). Calcifications were present in six patients, each with CDC. Clear tumor boundaries were visible in two patients with CDC and in nine with Xp11.2/TFE RCC (P < 0.05). The density of Xp11.2/TFE RCC tumors was greater than that of CDC tumors, normal renal cortex, or medulla on unenhanced CT. Enhancement was higher with Xp11.2/TFE RCC than with CDC tumors during all phases. Xp11.2/TFE RCC enhancement was higher than in the renal medulla during cortical and medullary phase but lower than in normal renal medulla during the delayed phase. CDC tumor enhancement was lower than that for normal renal medulla during all enhanced phases. CONCLUSION Both tumor types originated from the renal medulla. Distinguishing features included density on unenhanced CT, enhancement patterns, and capsule signs. Identifying these differences may aid diagnosis.
Collapse
Affiliation(s)
- Qing-Qiang Zhu
- Subei People's Hospital, Clinical School of Medical College, Yangzhou
| | - Zhong-Qiu Wang
- Department of Radiology, East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Rong Zhu
- Subei People's Hospital, Clinical School of Medical College, Yangzhou
| | - Wen-Xin Chen
- Subei People's Hospital, Clinical School of Medical College, Yangzhou
| | - Jing-Tao Wu
- Subei People's Hospital, Clinical School of Medical College, Yangzhou
| |
Collapse
|
22
|
Zhu Q, Wu J, Wang Z, Zhu W, Chen W, Wang S. WITHDRAWN: Comparative study of CT appearances in renal carcinoma associated with Xp11.2 translocation/TFE gene fusion and papillary renal cell carcinoma. Eur J Radiol 2012:S0720-048X(12)00550-5. [PMID: 23200629 DOI: 10.1016/j.ejrad.2012.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 09/16/2012] [Accepted: 11/01/2012] [Indexed: 12/01/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Qingqiang Zhu
- Department of Medical Imaging, Subei People's Hospital, Medical School of Yangzhou University, No. 98 West Nantong Road,Yangzhou 225001,China.
| | | | | | | | | | | |
Collapse
|
23
|
Klaassen Z, Tatem A, Burnette JO, Donohoe JM, Terris MK. Adult Xp11 translocation associated renal cell carcinoma: time to recognize. Urology 2012; 80:965-8. [PMID: 22999449 DOI: 10.1016/j.urology.2012.07.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 07/21/2012] [Accepted: 07/31/2012] [Indexed: 02/04/2023]
Affiliation(s)
- Zachary Klaassen
- Department of Surgery, Section of Urology, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | |
Collapse
|
24
|
Albiges L, Molinie V, Escudier B. Non-clear cell renal cell carcinoma: does the mammalian target of rapamycin represent a rational therapeutic target? Oncologist 2012; 17:1051-62. [PMID: 22807514 PMCID: PMC3425523 DOI: 10.1634/theoncologist.2012-0038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 06/20/2012] [Indexed: 12/19/2022] Open
Abstract
Non-clear cell renal cell carcinomas (nccRCCs) comprise a heterogenous and poorly characterized group of tumor types for which few treatments have been approved. Although targeted therapies have become the cornerstones of systemic treatment for metastatic renal cell carcinoma, patients with nccRCC have been excluded from many pivotal clinical trials. As such, robust clinical evidence supporting the use of these agents in patients with nccRCC is lacking. Here, we review the disparate nccRCC subtypes, the criteria for diagnosis, and the prognoses associated with each subtype, in addition to evaluating the potential use of mammalian target of rapamycin (mTOR) inhibitors in treating patients with nccRCC. Both genetic analyses and preclinical research indicate a central role for mTOR in nccRCC; a therapy that targets this ubiquitous regulator of cellular signaling could prove efficacious across various tumor subtypes. Results from recent studies exploring targeted therapies as both monotherapy and combination therapy have provided early indications of efficacy in patients with nccRCC. Exploratory analyses support further research with the mTOR inhibitors everolimus and temsirolimus in patients with nccRCC. Current clinical practice guidelines support the use of mTOR inhibitors in patients with nccRCC; however, these recommendations are based on low levels of evidence. Further results from randomized, controlled clinical trials are needed to determine the optimal choice of therapy for patients with nccRCC. Results from ongoing clinical trials of mTOR inhibitors and other agents in nccRCC, as well as their impact on the nccRCC treatment paradigm, are eagerly awaited.
Collapse
Affiliation(s)
- Laurence Albiges
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| | - Vincent Molinie
- Department of Pathology, Hôpital Saint Joseph, Paris, France
| | - Bernard Escudier
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|