1
|
Yoon AH, Tse JR. Hereditary renal mass syndromes: a pictorial review. Abdom Radiol (NY) 2024:10.1007/s00261-024-04534-y. [PMID: 39235599 DOI: 10.1007/s00261-024-04534-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/06/2024]
Abstract
Hereditary renal mass syndromes, although rare, account for at least 3-5% of kidney cancers and significantly impact affected families. Accurate diagnosis and management by radiologists are crucial as these syndromes often present at imaging with both renal and extra-renal manifestations. The radiologist may be the first to recognize these stigmata at imaging, some of which satisfy major features for diagnosis and obviate the need for genetic testing. Furthermore, radiologists contribute to lifelong imaging surveillance and locoregional treatment. This pictorial review discusses the following major hereditary renal mass syndromes with their typical renal mass appearance, extrarenal manifestations, inheritance pattern, diagnosis, and management strategies based on the most recent National Comprehensive Cancer Network guidelines: Von Hippel-Lindau syndrome, tuberous sclerosis complex, Birt-Hogg-Dube syndrome, hereditary paraganglioma/pheochromocytoma (PGL/PCC) syndrome/succinate dehydrogenase deficiency, hereditary leiomyomatosis and renal cell cancer (HLRCC)/fumarate hydratase deficiency, PTEN hamartoma syndrome, BRCA1-associated protein 1 (BAP1) tumor disposition syndrome, hereditary papillary renal cell carcinoma, and familial clear cell renal cell cancer with chromosome 3 translocation.
Collapse
Affiliation(s)
- Acacia H Yoon
- Menlo-Atherton High School, Atherton, CA, 94027, USA
| | - Justin R Tse
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
2
|
Alonso Grandes M, Roldán Testillano R, Márquez Negro A, Cernuda Pereira C, Dorado Valentín M, Khedaoui R, Páez Borda A. An atypical presentation of renal mass associated with BAP-1 tumor predisposition syndrome: Case report and review of literature. Urol Case Rep 2024; 54:102700. [PMID: 38827530 PMCID: PMC11143434 DOI: 10.1016/j.eucr.2024.102700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/18/2024] [Accepted: 02/27/2024] [Indexed: 06/04/2024] Open
Abstract
BCRA-associated protein-1 (BAP-1) mutation has been associated with the development of a familiar syndrome that predisposes to tumors with a higher incidence than in general population, including melanoma and renal carcinoma. We report a 47-year-old woman diagnosed with a BAPoma (melanocytic tumor characterized by the loss of BAP-1). Due to her extensive family history with multiple neoplasms, a FDG PET-CT was performed. Consequently, she was diagnosed with an atypical renal mass, which is rarely linked to this syndrome. We review and discuss the available literature on the screening, diagnosis and treatment of renal tumors associated with BAP-1 tumor predisposition syndrome.
Collapse
Affiliation(s)
- M. Alonso Grandes
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| | - R. Roldán Testillano
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| | - A.M. Márquez Negro
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| | - C. Cernuda Pereira
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| | - M. Dorado Valentín
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| | - R. Khedaoui
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
- University Hospital of Fuenlabrada. Department of Anatomical Pathology
| | - A. Páez Borda
- University Hospital of Fuenlabrada. Department of Urology. Fuenlabrada, Madrid, Spain
| |
Collapse
|
3
|
Waseh S, Lee JB. Advances in melanoma: epidemiology, diagnosis, and prognosis. Front Med (Lausanne) 2023; 10:1268479. [PMID: 38076247 PMCID: PMC10703395 DOI: 10.3389/fmed.2023.1268479] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/13/2023] [Indexed: 06/30/2024] Open
Abstract
Unraveling the multidimensional complexities of melanoma has required concerted efforts by dedicated community of researchers and clinicians battling against this deadly form of skin cancer. Remarkable advances have been made in the realm of epidemiology, classification, diagnosis, and therapy of melanoma. The treatment of advanced melanomas has entered the golden era as targeted personalized therapies have emerged that have significantly altered the mortality rate. A paradigm shift in the approach to melanoma classification, diagnosis, prognosis, and staging is underway, fueled by discoveries of genetic alterations in melanocytic neoplasms. A morphologic clinicopathologic classification of melanoma is expected to be replaced by a more precise molecular based one. As validated, convenient, and cost-effective molecular-based tests emerge, molecular diagnostics will play a greater role in the clinical and histologic diagnosis of melanoma. Artificial intelligence augmented clinical and histologic diagnosis of melanoma is expected to make the process more streamlined and efficient. A more accurate model of prognosis and staging of melanoma is emerging based on molecular understanding melanoma. This contribution summarizes the recent advances in melanoma epidemiology, classification, diagnosis, and prognosis.
Collapse
Affiliation(s)
- Shayan Waseh
- Department of Dermatology, Temple University Hospital, Philadelphia, PA, United States
| | - Jason B. Lee
- Department of Dermatology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
4
|
Shirole NH, Kaelin WG. von-Hippel Lindau and Hypoxia-Inducible Factor at the Center of Renal Cell Carcinoma Biology. Hematol Oncol Clin North Am 2023; 37:809-825. [PMID: 37270382 PMCID: PMC11315268 DOI: 10.1016/j.hoc.2023.04.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The most common form of kidney cancer is clear cell renal cell carcinoma (ccRCC). Biallelic VHL tumor suppressor gene inactivation is the usual initiating event in both hereditary (VHL Disease) and sporadic ccRCCs. The VHL protein, pVHL, earmarks the alpha subunits of the HIF transcription factor for destruction in an oxygen-dependent manner. Deregulation of HIF2 drives ccRCC pathogenesis. Drugs inhibiting the HIF2-responsive growth factor VEGF are now mainstays of ccRCC treatment. A first-in-class allosteric HIF2 inhibitor was recently approved for treating VHL Disease-associated neoplasms and appears active against sporadic ccRCC in early clinical trials.
Collapse
Affiliation(s)
- Nitin H Shirole
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - William G Kaelin
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Brigham and Women's Hospital, Harvard Medical School; Howard Hughes Medical Institute.
| |
Collapse
|
5
|
Szegedi K, Szabó Z, Kállai J, Király J, Szabó E, Bereczky Z, Juhász É, Dezső B, Szász C, Zsebik B, Flaskó T, Halmos G. Potential Role of VHL, PTEN, and BAP1 Mutations in Renal Tumors. J Clin Med 2023; 12:4538. [PMID: 37445575 DOI: 10.3390/jcm12134538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
The genetic profiling of renal tumors has revealed genomic regions commonly affected by structural changes and a general genetic heterogeneity. The VHL, PTEN, and BAP1 genes are often mutated in renal tumors. The frequency and clinical relevance of these mutations in renal tumors are still being researched. In our study, we investigated VHL, PTEN, and BAP1 genes and the sequencing of 24 samples of patients with renal tumors, revealing that VHL was mutated at a noticeable frequency (25%). Six of the investigated samples showed mutations, and one genetic polymorphism (rs779805) was detected in both heterozygote and homozygote forms. PTEN gene mutation was observed in only one sample, and one specimen showed genetic polymorphism. In the case of the BAP1 gene, all of the samples were wild types. Interestingly, VHL mutation was detected in two female patients diagnosed with AML and in one with oncocytoma. We assume that VHL or PTEN mutations may contribute to the development of human renal cancer. However, the overall mutation rate was low in all specimens investigated, and the development and prognosis of the disease were not exclusively associated with these types of genetic alterations.
Collapse
Affiliation(s)
- Krisztián Szegedi
- Department of Urology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Debrecen, 4032 Debrecen, Hungary
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Judit Kállai
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - József Király
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Erzsébet Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Juhász
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Balázs Dezső
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Oral Pathology and Microbiology, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - Csaba Szász
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Barbara Zsebik
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Tibor Flaskó
- Department of Urology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
6
|
Liu K, Huang Y, Xu Y, Wang G, Cai S, Zhang X, Shi T. BAP1-related signature predicts benefits from immunotherapy over VEGFR/mTOR inhibitors in ccRCC: a retrospective analysis of JAVELIN Renal 101 and checkmate-009/010/025 trials. Cancer Immunol Immunother 2023:10.1007/s00262-023-03424-4. [PMID: 37046008 DOI: 10.1007/s00262-023-03424-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND In patients with advanced clear cell renal cell carcinoma, despite the undoubted benefits from immune checkpoint inhibitor (ICI)-based therapies over monotherapies of angiogenic/mTOR inhibitors in the intention-to-treat population, approximately a quarter of the patients can scarcely gain advantage from ICIs, prompting the search for predictive biomarkers for patient selection. METHODS Clinical and multi-omic data of 2428 ccRCC patients were obtained from The Cancer Genome Atlas (TCGA, n = 537), JAVELIN Renal 101 (avelumab plus axitinib vs. sunitinib, n = 885), and CheckMate-009/010/025 (nivolumab vs. everolimus, n = 1006). RESULTS BAP1 mutations were associated with large progression-free survival (PFS) benefits from ICI-based immunotherapies over sunitinib/everolimus (pooled estimate of interaction HR = 0.71, 95% CI 0.51-0.99, P = 0.045). Using the top 20 BAP1 mutation-associated differentially expressed genes (DEGs) generated from the TCGA cohort, we developed the BAP1-score, negatively correlated with angiogenesis and positively correlated with multiple immune-related signatures concerning immune cell infiltration, antigen presentation, B/T cell receptor, interleukin, programmed death-1, and interferon. A high BAP1-score indicated remarkable PFS benefits from ICI-based immunotherapies over angiogenic/mTOR inhibitors (avelumab plus axitinib vs. sunitinib: HR = 0.55, 95% CI 0.43-0.70, P < 0.001; nivolumab vs. everolimus: HR = 0.72, 95% CI 0.52-1.00, P = 0.045), while these benefits were negligible in the low BAP1-score subgroup (HR = 1.16 and 1.02, respectively). CONCLUSION In advanced ccRCCs, the BAP1-score is a biologically and clinically significant predictor of immune microenvironment and the clinical benefits from ICI-based immunotherapies over angiogenic/mTOR inhibitors, demonstrating its potential utility in optimizing the personalized therapeutic strategies in patients with advanced ccRCC.
Collapse
Affiliation(s)
- Kan Liu
- Department of Urology, The Third Medical Center of PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Yan Huang
- Department of Urology, The Third Medical Center of PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | | | - Shangli Cai
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Xu Zhang
- Department of Urology, The Third Medical Center of PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| | - Taoping Shi
- Department of Urology, The Third Medical Center of PLA General Hospital, Yongding Road 69, Haidian District, Beijing, 100039, China.
| |
Collapse
|
7
|
Kwon J, Lee D, Lee SA. BAP1 as a guardian of genome stability: implications in human cancer. Exp Mol Med 2023; 55:745-754. [PMID: 37009801 PMCID: PMC10167335 DOI: 10.1038/s12276-023-00979-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/02/2023] [Accepted: 01/27/2023] [Indexed: 04/04/2023] Open
Abstract
BAP1 is a ubiquitin C-terminal hydrolase domain-containing deubiquitinase with a wide array of biological activities. Studies in which advanced sequencing technologies were used have uncovered a link between BAP1 and human cancer. Somatic and germline mutations of the BAP1 gene have been identified in multiple human cancers, with a particularly high frequency in mesothelioma, uveal melanoma and clear cell renal cell carcinoma. BAP1 cancer syndrome highlights that all carriers of inherited BAP1-inactivating mutations develop at least one and often multiple cancers with high penetrance during their lifetime. These findings, together with substantial evidence indicating the involvement of BAP1 in many cancer-related biological activities, strongly suggest that BAP1 functions as a tumor suppressor. Nonetheless, the mechanisms that account for the tumor suppressor function of BAP1 have only begun to be elucidated. Recently, the roles of BAP1 in genome stability and apoptosis have drawn considerable attention, and they are compelling candidates for key mechanistic factors. In this review, we focus on genome stability and summarize the details of the cellular and molecular functions of BAP1 in DNA repair and replication, which are crucial for genome integrity, and discuss the implications for BAP1-associated cancer and relevant therapeutic strategies. We also highlight some unresolved issues and potential future research directions.
Collapse
Affiliation(s)
- Jongbum Kwon
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea.
| | - Daye Lee
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
| | - Shin-Ai Lee
- Department of Life Science, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul, 03760, Korea
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Building 37, Room 1068, Bethesda, MD, 20892-4263, USA
| |
Collapse
|
8
|
Kapur P, Rajaram S, Brugarolas J. The expanding role of BAP1 in clear cell renal cell carcinoma. Hum Pathol 2023; 133:22-31. [PMID: 35932824 PMCID: PMC9898467 DOI: 10.1016/j.humpath.2022.07.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023]
Abstract
Mutations drive renal cell carcinoma biology and tumor growth. The BRCA1-associated protein-1 (BAP1) gene is frequently mutated in clear cell renal cell carcinoma (ccRCC) and has emerged as a prognostic and putative predictive biomarker. In this review, we discuss the role of BAP1 as a signature event of a subtype of ccRCC marked by aggressiveness, inflammation, and possibly a heightened response to immunotherapy.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA.
| | - Satwik Rajaram
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, Dallas, TX, 75390, USA; Department of Internal Medicine (Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
9
|
Webster BR, Gopal N, Ball MW. Tumorigenesis Mechanisms Found in Hereditary Renal Cell Carcinoma: A Review. Genes (Basel) 2022; 13:2122. [PMID: 36421797 PMCID: PMC9690265 DOI: 10.3390/genes13112122] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 09/29/2023] Open
Abstract
Renal cell carcinoma is a heterogenous cancer composed of an increasing number of unique subtypes each with their own cellular and tumor behavior. The study of hereditary renal cell carcinoma, which composes just 5% of all types of tumor cases, has allowed for the elucidation of subtype-specific tumorigenesis mechanisms that can also be applied to their sporadic counterparts. This review will focus on the major forms of hereditary renal cell carcinoma and the genetic alterations contributing to their tumorigenesis, including von Hippel Lindau syndrome, Hereditary Papillary Renal Cell Carcinoma, Succinate Dehydrogenase-Deficient Renal Cell Carcinoma, Hereditary Leiomyomatosis and Renal Cell Carcinoma, BRCA Associated Protein 1 Tumor Predisposition Syndrome, Tuberous Sclerosis, Birt-Hogg-Dubé Syndrome and Translocation RCC. The mechanisms for tumorigenesis described in this review are beginning to be exploited via the utilization of novel targets to treat renal cell carcinoma in a subtype-specific fashion.
Collapse
Affiliation(s)
| | | | - Mark W. Ball
- Center for Cancer Research, Urologic Oncology Branch, National Cancer Institute/NIH, 10 Center Drive, CRC Room 2W-5940, Bethesda, MD 20892, USA
| |
Collapse
|
10
|
Epidemiology and Prevention of Renal Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14164059. [PMID: 36011051 PMCID: PMC9406474 DOI: 10.3390/cancers14164059] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
With 400,000 diagnosed and 180,000 deaths in 2020, renal cell carcinoma (RCC) accounts for 2.4% of all cancer diagnoses worldwide. The highest disease burden developed countries, primarily in Europe and North America. Incidence is projected to increase in the future as more countries shift to Western lifestyles. Risk factors for RCC include fixed factors such as gender, age, and hereditary diseases, as well as intervening factors such as smoking, obesity, hypertension, diabetes, diet and alcohol, and occupational exposure. Intervening factors in primary prevention, understanding of congenital risk factors and the establishment of early diagnostic tools are important for RCC. This review will discuss RCC epidemiology, risk factors, and biomarkers involved in reducing incidence and improving survival.
Collapse
|
11
|
Xu DM, Li M, Lin SB, Yang ZL, Xu TY, Yang JH, Yin J. Comprehensive Analysis of Transcriptional Expression of hsa-mir-21 Predicted Target Genes and Immune Characteristics in Kidney Renal Clear Cell Carcinoma. Int J Med Sci 2022; 19:1482-1501. [PMID: 36035369 PMCID: PMC9413563 DOI: 10.7150/ijms.73404] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023] Open
Abstract
Background: To uncover advanced prognosis biomarkers in patient with kidney renal clear cell carcinoma (KIRC), our study was the first to make a comprehensive analysis of hsa-mir-21 predicted target genes and explore the immune characteristics in KIRC. Methods: In this study, the comprehensive analysis of hsa-mir-21 predicted target genes and immune characteristics in KIRC were analyzed via TIMER2.0, UALCAN, Metascape, Kaplan-Meier plotter, Human Protein Atlas, CancerSEA, JASPAR, GEPIA, R package: GSVA package (version 1.34.0) & immune infiltration algorithm (ssGSEA) and R package: RMS package (version 6.2-0) & SURVIVAL package (version 3.2-10). Results: Up-transcriptional expressions of RP2, NFIA, SPRY1 were significantly associated with favorable prognosis in KIRC, whereas that of TGFBI was markedly significantly to unfavorable prognosis. Additionally, RP2, NFIA, SPRY1 and TGFBI were significantly relevant to the immune infiltration in KIRC. Finally, ZNF263 was a common predicted transcription factor of RP2, NFIA, SPRY1 and TGFBI, which can as an independent indicator for prognosis in KIRC patients. Conclusions: Hsa-mir-21 predicted target genes (RP2, NFIA, SPRY1 and TGFBI) and the common transcription factor ZNF263 could be the advanced prognosis biomarkers in KIRC patients.
Collapse
Affiliation(s)
- Da-Ming Xu
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Ming Li
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Shu-Bin Lin
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Zheng-Liang Yang
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Teng-Yu Xu
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jin-Huan Yang
- Department of Urological Surgery, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jun Yin
- Department of Hematology, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
Vimercati L, Cavone D, Fortarezza F, Delfino MC, Ficarella R, Gentile A, De Palma A, Marulli G, De Maria L, Caporusso C, Marzullo A, d’Amati A, Romano DE, Caputi A, Sponselli S, Serio G, Pezzuto F. Case report: Mesothelioma and BAP1 tumor predisposition syndrome: Implications for public health. Front Oncol 2022; 12:966063. [PMID: 35992853 PMCID: PMC9386481 DOI: 10.3389/fonc.2022.966063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
BRCA-1 associated protein 1 (BAP1) tumour predisposition syndrome (TPDS) is a hereditary condition characterised by germline mutation of the tumour suppressor BAP1. This disorder is associated with the development of various benign and malignant tumours, mainly involving the skin, eyes, kidneys, and mesothelium. In this article, we report the case of a man recruited through the Apulia (Southern Italy) Mesothelioma Regional Operational Centre of the National Register of Mesotheliomas, who suffered from uveal melanoma, renal cancer, and mesothelioma, and a familial cluster of BAP1 germline mutations demonstrated by molecular analyses. The family members of the proband developed multiple malignancies. As tumours arising in this context have specific peculiarities in terms of clinical behaviour, identification of this condition through appropriate genetic counselling should be considered for adequate primary, secondary, and tertiary prevention measures for offspring.
Collapse
Affiliation(s)
- Luigi Vimercati
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Domenica Cavone
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Fortarezza
- Pathology Unit, Department of Medicine, School of Medicine and Surgery, University Hospital of Padova, University of Padova, Padova, Italy
| | - Maria Celeste Delfino
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Romina Ficarella
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Angela Gentile
- Medical Genetics Unit, Department of Human Reproductive Medicine, ASL Bari, Bari, Italy
| | - Angela De Palma
- Thoracic Surgery Unit, Department of Emergency and Organ Transplantation, University Hospital of Bari, Bari, Italy
| | - Giuseppe Marulli
- Thoracic Surgery Unit, Department of Emergency and Organ Transplantation, University Hospital of Bari, Bari, Italy
| | - Luigi De Maria
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Concetta Caporusso
- Department of Emergency and Organ Transplantation (DETO), Pathological Anatomy Section, University of Bari Aldo Moro, Bari, Italy
| | - Andrea Marzullo
- Department of Emergency and Organ Transplantation (DETO), Pathological Anatomy Section, University of Bari Aldo Moro, Bari, Italy
| | - Antonio d’Amati
- Department of Emergency and Organ Transplantation (DETO), Pathological Anatomy Section, University of Bari Aldo Moro, Bari, Italy
| | - Daniele Egidio Romano
- Department of Emergency and Organ Transplantation (DETO), Pathological Anatomy Section, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Caputi
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Stefania Sponselli
- Interdisciplinary Department of Medicine, Occupational Medicine Section Ramazzini, University of Bari Aldo Moro, Bari, Italy
| | - Gabriella Serio
- Department of Emergency and Organ Transplantation (DETO), Pathological Anatomy Section, University of Bari Aldo Moro, Bari, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health (DCTV), Pathology Unit, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Dawsey SJ, Gupta S. Hereditary Renal Cell Carcinoma. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Hereditary renal cell carcinoma (RCC) is a complex and rapidly evolving topic as there is a growing body of literature regarding inherited syndromes and mutations associated with an increased risk of RCC. OBJECTIVES: We sought to systematically review 13 hereditary syndromes associated with RCC; von Hippel-Lindau Disease associated RCC (VHLRCC), BAP-1 associated clear cell RCC (BAPccRCC), Familial non-von Hippel Lindau clear cell RCC (FccRCC), Tuberous Sclerosis Complex associated RCC (TSCRCC), Birt-Hogg-Dub e ´ Syndrome associated RCC (BHDRCC), PTEN Hamartoma Tumor Syndrome associated RCC (PHTSRCC), Microphthalmia-associated Transcription Family translocation RCC (MiTFtRCC), RCC with Chromosome 6p Amplification (TFEBRCC), Autosomal Dominant Polycystic Kidney Disease Associated RCC (ADPKDRCC), Hereditary Leiomyomatosis associated RCC (HLRCC), Succinate Dehydrogenase RCC (SDHRCC), Hereditary Papillary RCC (HPRCC), and ALK-Rearrangement RCC (ALKRCC). RESULTS: Hereditary RCC is generally associated with early age of onset, multifocal and/or bilateral lesions, and aggressive disease course. VHLRCC, BAPccRCC, FccRCC, and certain mutations resulting in SDHRCC are associated with clear cell RCC (ccRCC). HPRCC is associated with Type 1 papillary RCC. HLRCC is associated with type 2 papillary RCC. BHDRCC is associated with Chromophobe RCC. TSCRCC, PHTSRCC, MiTFtRCC, TFEBRCC, ADPKDRCC, certain SDHRCC and ALKRCC have variable histology. CONCLUSIONS: There has been tremendous advancement in our understanding of the pathophysiology of hereditary RCC. Ongoing research will refine our understanding of hereditary RCC and its therapeutic targets.
Collapse
Affiliation(s)
- Scott J. Dawsey
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Shilpa Gupta
- Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
14
|
Bell HN, Kumar-Sinha C, Mannan R, Zakalik D, Zhang Y, Mehra R, Jagtap D, Dhanasekaran SM, Vaishampayan U. Pathogenic ATM and BAP1 germline mutations in a case of early-onset, familial sarcomatoid renal cancer. Cold Spring Harb Mol Case Stud 2022; 8:mcs.a006203. [PMID: 35483881 PMCID: PMC9059789 DOI: 10.1101/mcs.a006203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/17/2022] [Indexed: 12/11/2022] Open
Abstract
Metastatic renal cell carcinoma (RCC) remains an incurable malignancy, despite recent advances in systemic therapies. Genetic syndromes associated with kidney cancer account for only 5%-8% of all diagnosed kidney malignancies, and genetic predispositions to kidney cancer predisposition are still being studied. Genomic testing for kidney cancer is useful for disease molecular subtyping but provides minimal therapeutic information. Understanding how aberrations drive RCC development and how their contextual influences, such as chromosome loss, genome instability, and DNA methylation changes, may alter therapeutic response is of importance. We report the case of a 36-yr-old female with aggressive, metastatic RCC and a significant family history of cancer, including RCC. This patient harbors a novel, pathogenic, germline ATM mutation along with a rare germline variant of unknown significance in the BAP1 gene. In addition, somatic loss of heterozygosity (LOH) in BAP1 and ATM genes, somatic mutation and LOH in the VHL gene, copy losses in Chromosomes 9p and 14, and genome instability are also noted in the tumor, potentially dictating this patient's aggressive clinical course. Further investigation is warranted to evaluate the association of ATM and BAP1 germline mutations with increased risk of RCC and if these mutations should lead to enhanced and early screening.
Collapse
Affiliation(s)
- Hannah N Bell
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Chandan Kumar-Sinha
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Rahul Mannan
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Dana Zakalik
- Oakland University/Beaumont Hospital, Rochester, Michigan 48309, USA
| | - Yuping Zhang
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Rohit Mehra
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Deepa Jagtap
- Oakland University/Beaumont Hospital, Rochester, Michigan 48309, USA
| | - Saravana M Dhanasekaran
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA.,Department of Pathology, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ulka Vaishampayan
- University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
15
|
Shao YF, DeBenedictis M, Yeaney G, Singh AD. Germ Line BAP1 Mutation in Patients with Uveal Melanoma and Renal Cell Carcinoma. Ocul Oncol Pathol 2021; 7:340-345. [PMID: 34722490 DOI: 10.1159/000516695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 04/17/2021] [Indexed: 12/19/2022] Open
Abstract
Uveal melanoma (UM) and renal cell carcinoma (RCC) can occur sporadically and as a manifestation of BAP1 tumor predisposition syndrome. We aimed to understand the prevalence of germ line BAP1 pathogenic variants in patients with UM and RCC. We reviewed patients managed at Cleveland Clinic between November 2003 and November 2019 who were diagnosed with UM and RCC. Charts were reviewed for demographic and cancer-related characteristics. RCC samples were tested for BAP1 protein expression using immunohistochemical (IHC) staining, and testing for germ line BAP1 pathogenic variants was performed as part of routine clinical care. Thirteen patients were included in the study. The average age at diagnosis of UM was 61.3 years. Seven patients underwent fine-needle aspiration biopsy for prognostic testing of UM (low risk =5, high risk =2). Twelve patients were treated with plaque radiation therapy, and 3 patients developed metastatic disease requiring systemic therapy. The median time to diagnosis of RCC from time of diagnosis of UM was 0 months. RCC samples were available for 7 patients for BAP1 IHC staining (intact =6, loss =1). All patients underwent nephrectomy (total = 3, partial = 8, unknown =2), and 1 received systemic therapy for metastatic RCC. Six patients underwent germ line BAP1 genetic testing. Of these, 1 patient was heterozygous for a pathogenic variant of BAP1 gene: c.1781-1782delGG, p.Gly594Valfs*48. The overall prevalence of germ line BAP1 pathogenic variants in our study was high (1/6; 17%; 95% CI 0-46%). Patients with UM and RCC should be referred for genetic counseling to discuss genetic testing.
Collapse
Affiliation(s)
- Yusra F Shao
- Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Gabrielle Yeaney
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Arun D Singh
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Gupta S, Erickson LA, Lohse CM, Shen W, Pitel BA, Knight SM, Halling KC, Herrera-Hernandez L, Boorjian SA, Thompson RH, Leibovich BC, Jimenez RE, Cheville JC. Assessment of Risk of Hereditary Predisposition in Patients With Melanoma and/or Mesothelioma and Renal Neoplasia. JAMA Netw Open 2021; 4:e2132615. [PMID: 34767027 PMCID: PMC8590170 DOI: 10.1001/jamanetworkopen.2021.32615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE In BAP1 tumor predisposition syndrome, clear cell renal cell carcinoma (RCC) is frequently associated with melanoma and/or mesothelioma, while germline MITF p.E318K alterations are being increasingly reported in melanoma/RCC. Limited data exist on the co-occurrence of melanoma and/or mesothelioma with renal neoplasia and the prevalence of associated germline alterations. OBJECTIVE To assess the frequency of melanoma and/or mesothelioma co-occurring with renal neoplasia using our institutional nephrectomy registry and to determine the prevalence of BAP1 and MITF alterations within this cohort. DESIGN, SETTING, AND PARTICIPANTS In this genetic association study, medical records from 8295 patients from 1970 to 2018, renal neoplasia co-occurring with melanoma and/or mesothelioma within a single institutional nephrectomy registry was reevaluated based on contemporary histopathologic criteria and the medical records were reviewed. Data were analyzed from September 2019 to May 2021. MAIN OUTCOMES AND MEASURES Identified cases were screened for BAP1 loss using immunohistochemistry; while patients with melanoma and clear cell RCC were screened for MITF p.E318K alterations. Tumors from patients with potential germline alterations were analyzed with comprehensive molecular profiling using a 514-gene next generation sequencing panel. RESULTS Of a total of 8295 patients, 93 (1.1%; 95% CI, 0.9%-1.4%) had melanoma and/or mesothelioma co-occurring with renal neoplasia (cutaneous melanoma, n = 76; uveal melanoma, n = 11; mesothelioma, n = 6). A total of 69 (74.2%) were male; 24 (25.8%) were female; median age at diagnosis of renal neoplasia was 63 years (IQR, 58-70 years) and the median duration of follow-up was 8.5 years (IQR, 5.0-14.6 years). Two patients with clear cell RCC had germline BAP1 alterations in the setting of cutaneous melanoma and mesothelioma. Two patients with hybrid oncocytic tumors had biallelic inactivation of FLCN in a setting of Birt-Hogg-Dubé (BHD) syndrome associated with uveal melanoma and mesothelioma. Tumor-only screening of clear cell RCC associated with cutaneous (n = 53) and uveal melanoma (n = 6) led to the identification of 1 patient with a likely germline MITF p.E318K alteration. After excluding benign renal neoplasia (such as oncocytoma and angiomyolipoma), alterations of BAP1, FLCN, and MITF were identified in 5 of 81 patients (6.2%) with melanoma and/or mesothelioma and renal neoplasia. In contrast to hybrid oncocytic tumors in BHD, no unique genotype-phenotype correlations were seen for clear cell RCC with pathogenic BAP1/ MITF alterations and VHL loss of function variants. Four of 5 cases (80%) met current National Comprehensive Cancer Network criteria for germline testing based on a combination of age, multifocality, histologic findings, and family history. CONCLUSIONS AND RELEVANCE In this genetic association study, findings support the continued use of these National Comprehensive Cancer Network criteria and suggest more stringent screening may be warranted in this patient population.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Lori A. Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Christine M. Lohse
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Wei Shen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Beth A. Pitel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Shannon M. Knight
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kevin C. Halling
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | - Rafael E. Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - John C. Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
17
|
Saly DL, Eswarappa MS, Street SE, Deshpande P. Renal Cell Cancer and Chronic Kidney Disease. Adv Chronic Kidney Dis 2021; 28:460-468.e1. [PMID: 35190112 DOI: 10.1053/j.ackd.2021.10.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/14/2021] [Accepted: 10/25/2021] [Indexed: 11/11/2022]
Abstract
The association between chronic kidney disease (CKD) and renal cell carcinoma (RCC) is bidirectional and multifactorial. Risk factors such as hypertension, diabetes mellitus, obesity, and smoking increase the risk of both CKD and RCC. CKD can lead to RCC via an underlying cystic disease or oxidative stress. RCC can cause CKD because of the tumor itself, surgical reduction of renal mass (either partial or radical nephrectomy), and perioperative acute kidney injury. Medical therapies such as immune checkpoint inhibitors and vascular endothelial growth factor inhibitors can lead to acute kidney injury and resultant CKD. Clinicians need to be aware of the complex, bidirectional interplay between both diseases.
Collapse
|
18
|
Gallan AJ, Parilla M, Segal J, Ritterhouse L, Antic T. BAP1-Mutated Clear Cell Renal Cell Carcinoma. Am J Clin Pathol 2021; 155:718-728. [PMID: 33210135 DOI: 10.1093/ajcp/aqaa176] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES While aberrations in the VHL gene and chromosome 3p resulting in clear cell renal cell carcinoma (CCRCC) are well established, we know that additional mutations in chromatin remodeling genes PBRM1, SETD2, and BRCA1-associated protein 1 (BAP1) contribute to pathogenesis in some cases. Given the known aggressive clinical behavior of BAP1-mutated CCRCC, we sought to define the pathologic phenotype of BAP1-mutated CCRCC. METHODS We identified 14 cases of molecularly proven BAP1-mutated CCRCC and investigated their clinicopathologic features. RESULTS BAP1-mutated CCRCC frequently showed papillary, tubulopapillary, or expanded nested architecture; demonstrated granular to diffusely eosinophilic cytoplasm with prominent eosinophilic globules; and contained high-grade nuclei. This morphology demonstrates significant overlap with Xp11 translocation renal cell carcinoma (RCC). Immunohistochemistry notably demonstrates loss of BAP1 expression in almost all tumors, in addition to strong p504S expression. A conventional CCRCC component was frequently present adjacent to the characteristic BAP1 areas and showed retained BAP1 expression and only patchy p504S. Approximately two-thirds of BAP1-mutated CCRCCs were stage pT3, renal vein invasion was common, and 50% developed metastases. CONCLUSIONS Herein, we describe the histologic and immunohistochemical findings in BAP1-mutated CCRCC, which has important implications for utilization of molecular testing, prognosis, future therapeutics, and distinction from other RCC subtypes such as Xp11 translocation RCC.
Collapse
Affiliation(s)
| | - Megan Parilla
- Department of Pathology, University of Chicago, Chicago, IL
| | - Jeremy Segal
- Department of Pathology, University of Chicago, Chicago, IL
| | | | - Tatjana Antic
- Department of Pathology, University of Chicago, Chicago, IL
| |
Collapse
|
19
|
Jonasch E, Walker CL, Rathmell WK. Clear cell renal cell carcinoma ontogeny and mechanisms of lethality. Nat Rev Nephrol 2021; 17:245-261. [PMID: 33144689 PMCID: PMC8172121 DOI: 10.1038/s41581-020-00359-2] [Citation(s) in RCA: 306] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 02/07/2023]
Abstract
The molecular features that define clear cell renal cell carcinoma (ccRCC) initiation and progression are being increasingly defined. The TRACERx Renal studies and others that have described the interaction between tumour genomics and remodelling of the tumour microenvironment provide important new insights into the molecular drivers underlying ccRCC ontogeny and progression. Our understanding of common genomic and chromosomal copy number abnormalities in ccRCC, including chromosome 3p loss, provides a mechanistic framework with which to organize these abnormalities into those that drive tumour initiation events, those that drive tumour progression and those that confer lethality. Truncal mutations in ccRCC, including those in VHL, SET2, PBRM1 and BAP1, may engender genomic instability and promote defects in DNA repair pathways. The molecular features that arise from these defects enable categorization of ccRCC into clinically and therapeutically relevant subtypes. Consideration of the interaction of these subtypes with the tumour microenvironment reveals that specific mutations seem to modulate immune cell populations in ccRCC tumours. These findings present opportunities for disease prevention, early detection, prognostication and treatment.
Collapse
Affiliation(s)
- Eric Jonasch
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Cheryl Lyn Walker
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
20
|
Tanaka T, Kawashima A, Marukawa Y, Kitayama T, Masaoka Y, Kojima K, Iguchi T, Hiraki T, Kanazawa S. Imaging evaluation of hereditary renal tumors: a pictorial review. Jpn J Radiol 2021; 39:619-632. [PMID: 33759057 DOI: 10.1007/s11604-021-01109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/10/2021] [Indexed: 11/28/2022]
Abstract
More than 10 hereditary renal tumor syndromes (HRTSs) and related germline mutations have been reported with HRTS-associated renal and extrarenal manifestations with benign and malignant tumors. Radiologists play an important role in detecting solitary or multiple renal masses with or without extrarenal findings on imaging and may raise the possibility of an inherited predisposition to renal cell carcinoma, providing direction for further screening, intervention and surveillance of the patients and their close family members before the development of potentially lethal renal and extrarenal tumors. Renal cell carcinomas (RCCs) associated with von Hippel-Lindau disease are typically slow growing while RCCs associated with HRTSs, such as hereditary leiomyomatosis and renal cell carcinoma syndrome, are highly aggressive. Therefore, radiologists need to be familiar with clinical and imaging findings of renal and extrarenal manifestations of HRTSs. This article reviews clinical and imaging findings for the evaluation of patients with well-established HRTSs from a radiologist's perspective to facilitate the clinical decision-making process for patient management.
Collapse
Affiliation(s)
- Takashi Tanaka
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan.
| | - Akira Kawashima
- Department of Radiology, Mayo Clinic, 13400 E Shea Blvd, Scottsdale, AZ, 85259, USA
| | - Yohei Marukawa
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Takahiro Kitayama
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Yoshihisa Masaoka
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Katsuhide Kojima
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Toshihiro Iguchi
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Takao Hiraki
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Susumu Kanazawa
- Department of Radiology, Okayama University Hospital, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| |
Collapse
|
21
|
Lang M, Vocke CD, Ricketts CJ, Metwalli AR, Ball MW, Schmidt LS, Linehan WM. Clinical and Molecular Characterization of Microphthalmia-associated Transcription Factor (MITF)-related Renal Cell Carcinoma. Urology 2021; 149:89-97. [PMID: 33242557 PMCID: PMC8728951 DOI: 10.1016/j.urology.2020.11.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To characterize the clinical presentation, genomic alterations, pathologic phenotype and clinical management of microphthalmia-associated transcription factor (MITF) familial renal cell carcinoma (RCC), caused by a member of the TFE3, TFEB, and MITF family of transcription factor genes. METHODS The clinical presentation, family history, tumor histopathology, and surgical management were evaluated and reported herein. DNA sequencing was performed on blood DNA, tumor DNA and DNA extracted from adjacent normal kidney tissue. Copy number and gene expression analyses on tumor and normal tissues were performed by Real-Time Polymerase chain reaction. TCGA gene expression data were used for comparative analysis. Protein expression and subcellular localization were evaluated by immunohistochemistry. RESULTS Germline genomic analysis identified the MITF p.E318K variant in a patient with bilateral, multifocal type 1 papillary RCC and a family history of RCC. All tumors displayed the MITF variant and were characterized by amplification of chromosomes 7 and 17, hallmarks of type 1 papillary RCC. We demonstrated that MITF p.E318K variant results in altered transcriptional activity and that downstream targets of MiT family members, such as GPNMB, are dysregulated in the tumors. CONCLUSION Association of the pathogenic MITF variant with bilateral and multifocal type 1 papillary RCC in this family supports its role as a risk allele for the development of RCC and emphasizes the importance of screening for MITF variants irrelevant of the RCC histologic subtype. This study identifies potential biomarkers for the disease, such as GPNMB expression, that may facilitate the development of targeted therapies for patients affected with MITF-associated RCC.
Collapse
Affiliation(s)
- Martin Lang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Cathy D Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Adam R Metwalli
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mark W Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Laura S Schmidt
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - William M Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
22
|
Gomella PT, Linehan WM, Ball MW. Precision Surgery and Kidney Cancer: Knowledge of Genetic Alterations Influences Surgical Management. Genes (Basel) 2021; 12:261. [PMID: 33670168 PMCID: PMC7916897 DOI: 10.3390/genes12020261] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/01/2021] [Accepted: 02/08/2021] [Indexed: 01/07/2023] Open
Abstract
Renal cell carcinoma is a term that represents multiple different disease processes, each driven by different genetic alterations, with distinct histology, and biological potential which necessitates divergent management strategies. This review discusses the genetic alterations seen in several forms of hereditary kidney cancer and how that knowledge can dictate when and how to intervene with a focus on the surgical management of these tumors.
Collapse
Affiliation(s)
| | | | - Mark W. Ball
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (P.T.G.); (W.M.L.)
| |
Collapse
|
23
|
Rong C, Zhou R, Wan S, Su D, Wang SL, Hess J. Ubiquitin Carboxyl-Terminal Hydrolases and Human Malignancies: The Novel Prognostic and Therapeutic Implications for Head and Neck Cancer. Front Oncol 2021; 10:592501. [PMID: 33585209 PMCID: PMC7878561 DOI: 10.3389/fonc.2020.592501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/07/2020] [Indexed: 12/24/2022] Open
Abstract
Ubiquitin C-terminal hydrolases (UCHs), a subfamily of deubiquitinating enzymes (DUBs), have been found in a variety of tumor entities and play distinct roles in the pathogenesis and development of various cancers including head and neck cancer (HNC). HNC is a heterogeneous disease arising from the mucosal epithelia of the upper aerodigestive tract, including different anatomic sites, distinct histopathologic types, as well as human papillomavirus (HPV)-positive and negative subgroups. Despite advances in multi-disciplinary treatment for HNC, the long-term survival rate of patients with HNC remains low. Emerging evidence has revealed the members of UCHs are associated with the pathogenesis and clinical prognosis of HNC, which highlights the prognostic and therapeutic implications of UCHs for patients with HNC. In this review, we summarize the physiological and pathological functions of the UCHs family, which provides enlightenment of potential mechanisms of UCHs family in HNC pathogenesis and highlights the potential consideration of UCHs as attractive drug targets.
Collapse
Affiliation(s)
- Chao Rong
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Ran Zhou
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shan Wan
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Dan Su
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Shou-Li Wang
- Department of Pathology, School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Jochen Hess
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Research Group Molecular Mechanisms of Head and Neck Tumors, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
24
|
Roles and mechanisms of BAP1 deubiquitinase in tumor suppression. Cell Death Differ 2021; 28:606-625. [PMID: 33462414 DOI: 10.1038/s41418-020-00709-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
The BAP1 gene has emerged as a major tumor suppressor mutated with various frequencies in numerous human malignancies, including uveal melanoma, malignant pleural mesothelioma, clear cell renal cell carcinoma, intrahepatic cholangiocarcinoma, hepatocellular carcinoma, and thymic epithelial tumors. BAP1 mutations are also observed at low frequency in other malignancies including breast, colorectal, pancreatic, and bladder cancers. BAP1 germline mutations are associated with high incidence of mesothelioma, uveal melanoma, and other cancers, defining the "BAP1 cancer syndrome." Interestingly, germline BAP1 mutations constitute an important paradigm for gene-environment interactions, as loss of BAP1 predisposes to carcinogen-induced tumorigenesis. Inactivating mutations of BAP1 are also identified in sporadic cancers, denoting the importance of this gene for normal tissue homeostasis and tumor suppression, although some oncogenic properties have also been attributed to BAP1. BAP1 belongs to the deubiquitinase superfamily of enzymes, which are responsible for the maturation and turnover of ubiquitin as well as the reversal of substrate ubiquitination, thus regulating ubiquitin signaling. BAP1 is predominantly nuclear and interacts with several chromatin-associated factors, assembling multi-protein complexes with mutually exclusive partners. BAP1 exerts its function through highly regulated deubiquitination of its substrates. As such, BAP1 orchestrates chromatin-associated processes including gene expression, DNA replication, and DNA repair. BAP1 also exerts cytoplasmic functions, notably in regulating Ca2+ signaling at the endoplasmic reticulum. This DUB is also subjected to multiple post-translational modifications, notably phosphorylation and ubiquitination, indicating that several signaling pathways tightly regulate its function. Recent progress indicated that BAP1 plays essential roles in multiple cellular processes including cell proliferation and differentiation, cell metabolism, as well as cell survival and death. In this review, we summarize the biological and molecular functions of BAP1 and explain how the inactivation of this DUB might cause human cancers. We also highlight some of the unresolved questions and suggest potential new directions.
Collapse
|
25
|
Carbone M, Arron ST, Beutler B, Bononi A, Cavenee W, Cleaver JE, Croce CM, D'Andrea A, Foulkes WD, Gaudino G, Groden JL, Henske EP, Hickson ID, Hwang PM, Kolodner RD, Mak TW, Malkin D, Monnat RJ, Novelli F, Pass HI, Petrini JH, Schmidt LS, Yang H. Tumour predisposition and cancer syndromes as models to study gene-environment interactions. Nat Rev Cancer 2020; 20:533-549. [PMID: 32472073 PMCID: PMC8104546 DOI: 10.1038/s41568-020-0265-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/23/2020] [Indexed: 12/18/2022]
Abstract
Cell division and organismal development are exquisitely orchestrated and regulated processes. The dysregulation of the molecular mechanisms underlying these processes may cause cancer, a consequence of cell-intrinsic and/or cell-extrinsic events. Cellular DNA can be damaged by spontaneous hydrolysis, reactive oxygen species, aberrant cellular metabolism or other perturbations that cause DNA damage. Moreover, several environmental factors may damage the DNA, alter cellular metabolism or affect the ability of cells to interact with their microenvironment. While some environmental factors are well established as carcinogens, there remains a large knowledge gap of others owing to the difficulty in identifying them because of the typically long interval between carcinogen exposure and cancer diagnosis. DNA damage increases in cells harbouring mutations that impair their ability to correctly repair the DNA. Tumour predisposition syndromes in which cancers arise at an accelerated rate and in different organs - the equivalent of a sensitized background - provide a unique opportunity to examine how gene-environment interactions influence cancer risk when the initiating genetic defect responsible for malignancy is known. Understanding the molecular processes that are altered by specific germline mutations, environmental exposures and related mechanisms that promote cancer will allow the design of novel and effective preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA.
| | - Sarah T Arron
- STA, JEC, Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce Beutler
- Center for Genetic Host Defense, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Angela Bononi
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Webster Cavenee
- Ludwig Institute, University of California, San Diego, San Diego, CA, USA
| | - James E Cleaver
- STA, JEC, Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, Ohio State University, Columbus, OH, USA
| | - Alan D'Andrea
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Giovanni Gaudino
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Elizabeth P Henske
- Center for LAM Research, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ian D Hickson
- Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Paul M Hwang
- Cardiovascular Branch, National Institutes of Health, Bethesda, MD, USA
| | - Richard D Kolodner
- Ludwig Institute, University of California, San Diego, San Diego, CA, USA
| | - Tak W Mak
- Princess Margaret Cancer Center, University of Toronto, Toronto, ON, Canada
| | - David Malkin
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Raymond J Monnat
- Department Pathology, Washington University, Seattle, WA, USA
- Department of Genome Science, Washington University, Seattle, WA, USA
| | - Flavia Novelli
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Harvey I Pass
- Department of Cardiovascular Surgery, New York University, New York, NY, USA
| | - John H Petrini
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura S Schmidt
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI, USA
| |
Collapse
|
26
|
Testa U, Pelosi E, Castelli G. Genetic Alterations in Renal Cancers: Identification of The Mechanisms Underlying Cancer Initiation and Progression and of Therapeutic Targets. MEDICINES (BASEL, SWITZERLAND) 2020; 7:E44. [PMID: 32751108 PMCID: PMC7459851 DOI: 10.3390/medicines7080044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/19/2020] [Accepted: 07/24/2020] [Indexed: 12/26/2022]
Abstract
Renal cell cancer (RCC) involves three most recurrent sporadic types: clear-cell RCC (70-75%, CCRCC), papillary RCCC (10-15%, PRCC), and chromophobe RCC (5%, CHRCC). Hereditary cases account for about 5% of all cases of RCC and are caused by germline pathogenic variants. Herein, we review how a better understanding of the molecular biology of RCCs has driven the inception of new diagnostic and therapeutic approaches. Genomic research has identified relevant genetic alterations associated with each RCC subtype. Molecular studies have clearly shown that CCRCC is universally initiated by Von Hippel Lindau (VHL) gene dysregulation, followed by different types of additional genetic events involving epigenetic regulatory genes, dictating disease progression, aggressiveness, and differential response to treatments. The understanding of the molecular mechanisms that underlie the development and progression of RCC has considerably expanded treatment options; genomic data might guide treatment options by enabling patients to be matched with therapeutics that specifically target the genetic alterations present in their tumors. These new targeted treatments have led to a moderate improvement of the survival of metastatic RCC patients. Ongoing studies based on the combination of immunotherapeutic agents (immune check inhibitors) with VEGF inhibitors are expected to further improve the survival of these patients.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Vaile Regina Elena 299, 00161 Rome, Italy; (E.P.); (G.C.)
| | | | | |
Collapse
|
27
|
Carbone M, Harbour JW, Brugarolas J, Bononi A, Pagano I, Dey A, Krausz T, Pass HI, Yang H, Gaudino G. Biological Mechanisms and Clinical Significance of BAP1 Mutations in Human Cancer. Cancer Discov 2020; 10:1103-1120. [PMID: 32690542 DOI: 10.1158/2159-8290.cd-19-1220] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/03/2020] [Accepted: 05/07/2020] [Indexed: 11/16/2022]
Abstract
Among more than 200 BAP1-mutant families affected by the "BAP1 cancer syndrome," nearly all individuals inheriting a BAP1 mutant allele developed one or more malignancies during their lifetime, mostly uveal and cutaneous melanoma, mesothelioma, and clear-cell renal cell carcinoma. These cancer types are also those that, when they occur sporadically, are more likely to carry somatic biallelic BAP1 mutations. Mechanistic studies revealed that the tumor suppressor function of BAP1 is linked to its dual activity in the nucleus, where it is implicated in a variety of processes including DNA repair and transcription, and in the cytoplasm, where it regulates cell death and mitochondrial metabolism. BAP1 activity in tumor suppression is cell type- and context-dependent. BAP1 has emerged as a critical tumor suppressor across multiple cancer types, predisposing to tumor development when mutated in the germline as well as somatically. Moreover, BAP1 has emerged as a key regulator of gene-environment interaction.This article is highlighted in the In This Issue feature, p. 1079.
Collapse
Affiliation(s)
| | - J William Harbour
- Bascom Palmer Eye Institute, Sylvester Comprehensive Cancer Center, and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Angela Bononi
- University of Hawai'i Cancer Center, Honolulu, Hawai'i
| | - Ian Pagano
- University of Hawai'i Cancer Center, Honolulu, Hawai'i
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, South San Francisco, California
| | - Thomas Krausz
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York
| | - Haining Yang
- University of Hawai'i Cancer Center, Honolulu, Hawai'i
| | | |
Collapse
|
28
|
Ball MW, An JY, Gomella PT, Gautam R, Ricketts CJ, Vocke CD, Schmidt LS, Merino MJ, Srinivasan R, Malayeri AA, Metwalli AR, Linehan WM. Growth Rates of Genetically Defined Renal Tumors: Implications for Active Surveillance and Intervention. J Clin Oncol 2020; 38:1146-1153. [PMID: 32083993 PMCID: PMC7145590 DOI: 10.1200/jco.19.02263] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2020] [Indexed: 01/10/2023] Open
Abstract
PURPOSE Published series of growth rates of renal tumors on active surveillance largely consist of tumors without pathologic or genetic data. Growth kinetics of genetically defined renal tumors are not well known. Here, we evaluate the growth of genetically defined renal tumors and their association with patient clinical and genetic characteristics. PATIENTS AND METHODS We evaluated patients with an inherited kidney cancer susceptibility syndrome as a result of a pathologic germline alteration of VHL, MET, FLCN, or BAP1 with at least 1 solid renal mass managed with active surveillance at our institution. Tumor growth rates (GR) were calculated and patients were stratified by genetic alteration and other clinical and genetic factors to analyze differences in growth rates using linear regression and comparative statistics. RESULTS A total of 292 patients with 435 genetically defined tumors were identified, including 286 VHL-deficient, 91 FLCN-deficient, 52 MET-activated, and 6 BAP1-deficient tumors. There were significant differences in GRs when stratified by genetic alteration. BAP1-deficient tumors had the fastest median GR (0.6 cm/y; interquartile range [IQR], 0.57-0.68 cm/y), followed by VHL-deficient tumors (GR, 0.37 cm/y; IQR, 0.25-0.57 cm/y), FLCN-deficient tumors (GR, 0.10 cm/y; IQR, 0.04-0.24 cm/y), and tumors with MET activation (GR, 0.15 cm/y; IQR, 0.053-0.32 cm/y; P < .001). Tumors from the same patient had similar GRs. Younger age was independently associated with higher GR (P = .005). CONCLUSION In a cohort of genetically defined tumors, tumor growth rates varied in a clinically and statistically different manner according to genetic subtype. Rapid growth of BAP1-deficient tumors indicates that these patients should be managed with caution. The faster growth of tumors in younger patients may support more frequent imaging, whereas the slower growth of other tumors may support extended surveillance beyond annual imaging in some instances.
Collapse
Affiliation(s)
- Mark W. Ball
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Julie Y. An
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Patrick T. Gomella
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rabindra Gautam
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Christopher J. Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Cathy D. Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Laura S. Schmidt
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Maria J. Merino
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ramaprasad Srinivasan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Ashkan A. Malayeri
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Adam R. Metwalli
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - W. Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW To describe current paradigms for genetic testing, screening, and treatment of patients with inherited kidney cancer syndromes. RECENT FINDINGS We describe various new aspects of hereditary kidney cancer. Recent data now support that hereditary kidney cancer may account for 5-8% of kidney cancers diagnosed. Methods of testing have evolved including the introduction of multigene next-generation sequencing panels. We continue to learn more about the natural history and management of classic hereditary cancer syndromes. New emerging conditions with lower kidney cancer penetrance have been recognized adding the growing list of syndromes associated with kidney cancer development. The surgical management strategies of enucleation remain however systemic therapy options are being explored both for localized and advanced settings. SUMMARY Genetic predisposition to kidney cancer is likely more common than once thought. Knowledge of clinical manifestation and genetic testing strategies are needed to properly identify and treat patient and their families.
Collapse
|
30
|
BAP1 Loss is a Useful Adjunct to Distinguish Malignant Mesothelioma Including the Adenomatoid-like Variant From Benign Adenomatoid Tumors. Appl Immunohistochem Mol Morphol 2020; 28:67-73. [DOI: 10.1097/pai.0000000000000700] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
31
|
Carlo MI, Hakimi AA, Stewart GD, Bratslavsky G, Brugarolas J, Chen YB, Linehan WM, Maher ER, Merino MJ, Offit K, Reuter VE, Shuch B, Coleman JA. Familial Kidney Cancer: Implications of New Syndromes and Molecular Insights. Eur Urol 2019; 76:754-764. [PMID: 31326218 PMCID: PMC7673107 DOI: 10.1016/j.eururo.2019.06.015] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/12/2019] [Indexed: 02/07/2023]
Abstract
CONTEXT Hereditary cases account for about 5% of all cases of renal cell carcinoma (RCC). With advances in next-generation sequencing, several new hereditary syndromes have been described in the last few years. OBJECTIVE To review and summarise the recent preclinical and clinical literature in hereditary renal cancer. EVIDENCE ACQUISITION A systematic review of the literature was performed in November 2018 using PubMed and OMIM databases, with an emphasis on kidney cancer, genetics and genomics, clinical criteria, and management. EVIDENCE SYNTHESIS Several autosomal dominant hereditary RCC syndromes have been described, including those related to germline pathogenic variants in VHL, MET, FH, TSC1/TSC2, FLCN, SDHA/B/C/D, BAP1, CDC73, and MITF. Clinical spectrum of SDH, BAP1, and MITF is still being defined, although these appear to be associated with a lower incidence of RCC. FH and likely BAP1 RCC are associated with more aggressive disease. Preclinical and clinical studies show that using systemic therapy that exploits specific genetic pathways is a promising strategy. CONCLUSIONS There are several well-described hereditary RCC syndromes, as well as recently identified ones, for which the full clinical spectrum is yet to be defined. In the new era of precision medicine, identification of these syndromes may play an important role in management and systemic treatment selection. PATIENT SUMMARY This review covers updates in the diagnosis and management of familial kidney cancer syndromes. We describe updates in testing and management of the most common syndromes such as von Hippel-Lindau, and hereditary leiomyomatosis and renal cell carcinoma. We also provide insights into recently described familial kidney cancer syndromes.
Collapse
Affiliation(s)
- Maria I Carlo
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - A Ari Hakimi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Ying-Bei Chen
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - W Marston Linehan
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eamonn R Maher
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Cenre, Cambridge, UK
| | - Maria J Merino
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Brian Shuch
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
32
|
Machida YJ. A mechanism for the tissue specificity in BAP1 cancer syndrome. Transl Cancer Res 2019; 8:S621-S624. [PMID: 35117145 PMCID: PMC8798120 DOI: 10.21037/tcr.2019.06.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 06/25/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Yuichi J Machida
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
33
|
Imaging surveillance for children with predisposition to renal tumors. Pediatr Radiol 2019; 49:1453-1462. [PMID: 31620846 DOI: 10.1007/s00247-019-04432-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/08/2019] [Accepted: 05/15/2019] [Indexed: 12/16/2022]
Abstract
Effective surveillance is necessary for early detection of tumors in children with cancer predisposition syndromes. Instituting a surveillance regimen in children comes with practical challenges that include determining imaging modality and timing, and considering cost efficiency, accessibility, and the significant consequences of false-positive and false-negative results. To address these challenges, the American Association for Cancer Research has recently published consensus recommendations that focus on surveillance of cancer predisposition syndromes in children. This review condenses the imaging surveillance recommendations for syndromes that carry a predisposition to renal tumors in childhood, and includes summaries of the predisposition syndromes and discussion of considerations of available imaging modalities.
Collapse
|
34
|
Chau C, van Doorn R, van Poppelen NM, van der Stoep N, Mensenkamp AR, Sijmons RH, van Paassen BW, van den Ouweland AMW, Naus NC, van der Hout AH, Potjer TP, Bleeker FE, Wevers MR, van Hest LP, Jongmans MCJ, Marinkovic M, Bleeker JC, Jager MJ, Luyten GPM, Nielsen M. Families with BAP1-Tumor Predisposition Syndrome in The Netherlands: Path to Identification and a Proposal for Genetic Screening Guidelines. Cancers (Basel) 2019; 11:cancers11081114. [PMID: 31382694 PMCID: PMC6721807 DOI: 10.3390/cancers11081114] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/26/2019] [Accepted: 08/01/2019] [Indexed: 12/26/2022] Open
Abstract
Germline pathogenic variants in the BRCA1-associated protein-1 (BAP1) gene cause the BAP1-tumor predisposition syndrome (BAP1-TPDS, OMIM 614327). BAP1-TPDS is associated with an increased risk of developing uveal melanoma (UM), cutaneous melanoma (CM), malignant mesothelioma (MMe), renal cell carcinoma (RCC), meningioma, cholangiocarcinoma, multiple non-melanoma skin cancers, and BAP1-inactivated nevi. Because of this increased risk, it is important to identify patients with BAP1-TPDS. The associated tumors are treated by different medical disciplines, emphasizing the need for generally applicable guidelines for initiating genetic analysis. In this study, we describe the path to identification of BAP1-TPDS in 21 probands found in the Netherlands and the family history at the time of presentation. We report two cases of de novo BAP1 germline mutations (2/21, 9.5%). Findings of this study combined with previously published literature, led to a proposal of guidelines for genetic referral. We recommend genetic analysis in patients with ≥2 BAP1-TPDS-associated tumors in their medical history and/or family history. We also propose to test germline BAP1 in patients diagnosed with UM <40 years, CM <18 years, MMe <50 years, or RCC <46 years. Furthermore, other candidate susceptibility genes for tumor types associated with BAP1-TPDS are discussed, which can be included in gene panels when testing patients.
Collapse
Affiliation(s)
- Cindy Chau
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Remco van Doorn
- Department of Dermatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Natasha M van Poppelen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Department of Ophthalmology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Nienke van der Stoep
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Arjen R Mensenkamp
- Department of Clinical Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Rolf H Sijmons
- Department of Genetics, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Barbara W van Paassen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Nicole C Naus
- Department of Ophthalmology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | | | - Thomas P Potjer
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Fonnet E Bleeker
- Department of Clinical Genetics, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
| | - Marijke R Wevers
- Department of Clinical Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Liselotte P van Hest
- Department of Clinical Genetics, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
| | - Marjolijn C J Jongmans
- Department of Clinical Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Department of Clinical Genetics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Marina Marinkovic
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaco C Bleeker
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gregorius P M Luyten
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Maartje Nielsen
- Department of Clinical Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
35
|
Snow A, Ricker C, In GK. Two synchronous malignancies: nodular melanoma and renal cell carcinoma in a patient with an underlying germline BRCA2 mutation. BMJ Case Rep 2019; 12:12/6/e227625. [PMID: 31227566 DOI: 10.1136/bcr-2018-227625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Modernised genetic testing among patients with cancer has led to an increasing wealth of knowledge regarding cancer biology and aetiology. Furthermore, some germline mutations have the potential to direct therapeutic approaches as well. While BRCA1/2 mutations are well-established risk factors for breast and ovarian cancers, their impact on other cancers is less understood. We describe a patient with a germline BRCA2 mutation who developed synchronous melanoma and renal cell carcinoma, but responded well to treatment and is now cancer free.
Collapse
Affiliation(s)
- Anson Snow
- Department of Medicine, Los Angeles County University of Southern California Medical Center, Los Angeles, California, USA
| | - Charite Ricker
- Department of Medicine, Division of Medical Oncology, University of Southern California - Norris Comprehensive Cancer Center, Los Angeles, California, USA
| | - Gino K In
- Department of Dermatology, University of Southern California - Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
36
|
Christensen MB, Wadt K, Jensen UB, Lautrup CK, Bojesen A, Krogh LN, van Overeem Hansen T, Gerdes AM. Exploring the hereditary background of renal cancer in Denmark. PLoS One 2019; 14:e0215725. [PMID: 31034483 PMCID: PMC6488054 DOI: 10.1371/journal.pone.0215725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Every year more than 800 patients in Denmark are diagnosed with renal cell carcinoma (RCC) of which 3-5% are expected to be part of a hereditary renal cancer syndrome. We performed genetic screening of causative and putative RCC-genes (VHL, FH, FLCN, MET, SDHB, BAP1, MITF, CDKN2B) in RCC-patients suspected of a genetic predisposition. METHODS The cohort consisted of forty-eight Danish families or individuals with early onset RCC, a family history of RCC, a family history of RCC and melanoma or both RCC- and melanoma diagnosis in the same individual. DNA was extracted from peripheral blood samples or cancer-free formalin-fixed paraffin-embedded tissue. RESULTS One start codon variant of unknown clinical significance (VUS) (c.3G>A, p.Met1Ile) and one missense VUS (c.631A>C, p.Met211Leu) was found in VHL in a patient with RCC-onset at twenty-eight years of age but without other manifestations or family history of von Hippel-Lindau (VHL). Furthermore, in three families we found three different variants in BAP1, one of which was a novel non-segregating missense variant (c.1502G>A, p.Ser501Asn) in a family with two brothers affected with RCC. Finally, we found the known E318K-substitution in MITF in a RCC-affected member of a family with multiple melanomas. No variants were detected in CDKN2B. CONCLUSION Although we did find three VUS's in BAP1 in three families and a pathogenic variant in MITF in one family, pathogenic germline variants in BAP1, MITF or CDKN2B are not frequent causes of hereditary renal cancer in Denmark. It is possible that the high prevalence of risk factors such as male gender, smoking and obesity has influenced the development of cancer in the patients of the current study. Further investigations into putative predisposing genes and risk factors of RCC are necessary to enable better prediction of renal cancer risk or presymptomatic testing of relatives in hereditary renal cancer families.
Collapse
Affiliation(s)
| | - Karin Wadt
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
| | - Uffe Birk Jensen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| | | | - Anders Bojesen
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Genetics, Sygehus Lillebaelt, Vejle, Denmark
| | | | | | - Anne-Marie Gerdes
- Department of Clinical Genetics, Copenhagen University Hospital, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
37
|
El Bouchtaoui M, Do Cruzeiro M, Leboeuf C, Loisel-Ferreira I, Fedronie C, Ferreira C, Ait El Far R, Ziol M, Espié M, Falgarone G, Cassinat B, Kiladjian JJ, Feugeas JP, Janin A, Bousquet G. A Constitutional Activating MET Mutation Makes the Genetic Link between Malignancies and Chronic Inflammatory Diseases. Clin Cancer Res 2019; 25:4504-4515. [DOI: 10.1158/1078-0432.ccr-18-3261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/16/2019] [Accepted: 04/15/2019] [Indexed: 11/16/2022]
|
38
|
He M, Chaurushiya MS, Webster JD, Kummerfeld S, Reja R, Chaudhuri S, Chen YJ, Modrusan Z, Haley B, Dugger DL, Eastham-Anderson J, Lau S, Dey A, Caothien R, Roose-Girma M, Newton K, Dixit VM. Intrinsic apoptosis shapes the tumor spectrum linked to inactivation of the deubiquitinase BAP1. SCIENCE (NEW YORK, N.Y.) 2019; 364:283-285. [PMID: 31000662 DOI: 10.1126/science.aav4902] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/15/2019] [Indexed: 11/02/2022]
Abstract
Malignancies arising from mutation of tumor suppressors have unexplained tissue proclivity. For example, BAP1 encodes a widely expressed deubiquitinase for histone H2A, but germline mutations are predominantly associated with uveal melanomas and mesotheliomas. We show that BAP1 inactivation causes apoptosis in mouse embryonic stem cells, fibroblasts, liver, and pancreatic tissue but not in melanocytes and mesothelial cells. Ubiquitin ligase RNF2, which silences genes by monoubiquitinating H2A, promoted apoptosis in BAP1-deficient cells by suppressing expression of the prosurvival genes Bcl2 and Mcl1. In contrast, BAP1 loss in melanocytes had little impact on expression of prosurvival genes, instead inducing Mitf Thus, BAP1 appears to modulate gene expression by countering H2A ubiquitination, but its loss only promotes tumorigenesis in cells that do not engage an RNF2-dependent apoptotic program.
Collapse
Affiliation(s)
- Meng He
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mira S Chaurushiya
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joshua D Webster
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Sarah Kummerfeld
- Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Rohit Reja
- Department of Bioinformatics, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Subhra Chaudhuri
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ying-Jiun Chen
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zora Modrusan
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin Haley
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Debra L Dugger
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Shari Lau
- Department of Pathology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Anwesha Dey
- Department of Discovery Oncology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Roger Caothien
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Merone Roose-Girma
- Department of Molecular Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kim Newton
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Vishva M Dixit
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
39
|
Perera RM, Di Malta C, Ballabio A. MiT/TFE Family of Transcription Factors, Lysosomes, and Cancer. ANNUAL REVIEW OF CANCER BIOLOGY 2019; 3:203-222. [PMID: 31650096 PMCID: PMC6812561 DOI: 10.1146/annurev-cancerbio-030518-055835] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer cells have an increased demand for energy sources to support accelerated rates of growth. When nutrients become limiting, cancer cells may switch to nonconventional energy sources that are mobilized through nutrient scavenging pathways involving autophagy and the lysosome. Thus, several cancers are highly reliant on constitutive activation of these pathways to degrade and recycle cellular materials. Here, we focus on the MiT/TFE family of transcription factors, which control transcriptional programs for autophagy and lysosome biogenesis and have emerged as regulators of energy metabolism in cancer. These new findings complement earlier reports that chromosomal translocations and amplifications involving the MiT/TFE genes contribute to the etiology and pathophysiology of renal cell carcinoma, melanoma, and sarcoma, suggesting pleiotropic roles for these factors in a wider array of cancers. Understanding the interplay between the oncogenic and stress-adaptive roles of MiT/TFE factors could shed light on fundamental mechanisms of cellular homeostasis and point to new strategies for cancer treatment.
Collapse
Affiliation(s)
- Rushika M Perera
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, 80138Naples, Italy
- Department of Molecular and Human Genetics and Neurological Research Institute, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
40
|
Wu J, Wang H, Ricketts CJ, Yang Y, Merino MJ, Zhang H, Shi G, Gan H, Linehan WM, Zhu Y, Ye D. Germline mutations of renal cancer predisposition genes and clinical relevance in Chinese patients with sporadic, early-onset disease. Cancer 2018; 125:1060-1069. [PMID: 30548481 DOI: 10.1002/cncr.31908] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/04/2018] [Accepted: 11/12/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND An inherited susceptibility to renal cancers is associated with multiple predisposing genes, but most screening tests are limited to patients with a family history. Next-generation sequencing (NGS)-based multigene panels provide an efficient and adaptable tool for investigating pathogenic germline mutations on a larger scale. This study investigated the frequency of pathogenic germline mutations in renal cancer predisposition genes in patients with sporadic, early-onset disease. METHODS An NGS-based panel of 23 known and potential renal cancer predisposition genes was used to analyze germline mutations in 190 unrelated Chinese patients under the age of 45 years who presented with renal tumors. The detected variants were filtered for pathogenicity, and then their frequencies were calculated and correlated with clinical features. Germline variants of the fumarate hydratase (FH) and BRCA1-associated protein 1 (BAP1) genes were comprehensively analyzed because of their aggressive potential. RESULTS In total, 18 patients (9.5%) had germline mutations in 10 genes. Twelve of these 18 patients had alterations in renal cancer predisposition genes (6.3%), and 6 patients had mutations in potential predisposition genes such as BRCA1/2. Notably, pathogenic mutation carriers had a significant family history in second-degree relatives in comparison with those without pathogenic mutations (P < .001). Variants of unknown clinical significance in FH and BAP1 demonstrated evidence of additional somatic loss in tumors. CONCLUSIONS In patients with early-onset disease, a multigene panel identified a high pathogenic germline mutation rate in renal cancer predisposition genes. This study emphasizes the importance of screening patients with early-onset disease for mutations in cancer predisposition genes. Germline screening should be encouraged in early-onset patients to provide personalized medicine and improve patient outcomes.
Collapse
Affiliation(s)
- Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Hongkai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Christopher J Ricketts
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Youfeng Yang
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria J Merino
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China
| | - W Marston Linehan
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
41
|
Walpole S, Pritchard AL, Cebulla CM, Pilarski R, Stautberg M, Davidorf FH, de la Fouchardière A, Cabaret O, Golmard L, Stoppa-Lyonnet D, Garfield E, Njauw CN, Cheung M, Turunen JA, Repo P, Järvinen RS, van Doorn R, Jager MJ, Luyten GPM, Marinkovic M, Chau C, Potrony M, Höiom V, Helgadottir H, Pastorino L, Bruno W, Andreotti V, Dalmasso B, Ciccarese G, Queirolo P, Mastracci L, Wadt K, Kiilgaard JF, Speicher MR, van Poppelen N, Kilic E, Al-Jamal RT, Dianzani I, Betti M, Bergmann C, Santagata S, Dahiya S, Taibjee S, Burke J, Poplawski N, O’Shea SJ, Newton-Bishop J, Adlard J, Adams DJ, Lane AM, Kim I, Klebe S, Racher H, Harbour JW, Nickerson ML, Murali R, Palmer JM, Howlie M, Symmons J, Hamilton H, Warrier S, Glasson W, Johansson P, Robles-Espinoza CD, Ossio R, de Klein A, Puig S, Ghiorzo P, Nielsen M, Kivelä TT, Tsao H, Testa JR, Gerami P, Stern MH, Paillerets BBD, Abdel-Rahman MH, Hayward NK. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide. J Natl Cancer Inst 2018; 110:1328-1341. [PMID: 30517737 PMCID: PMC6292796 DOI: 10.1093/jnci/djy171] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 07/17/2018] [Accepted: 08/31/2018] [Indexed: 12/17/2022] Open
Abstract
Background The BRCA1-associated protein-1 (BAP1) tumor predisposition syndrome (BAP1-TPDS) is a hereditary tumor syndrome caused by germline pathogenic variants in BAP1 encoding a tumor suppressor associated with uveal melanoma, mesothelioma, cutaneous melanoma, renal cell carcinoma, and cutaneous BAP1-inactivated melanocytic tumors. However, the full spectrum of tumors associated with the syndrome is yet to be determined. Improved understanding of the BAP1-TPDS is crucial for appropriate clinical management of BAP1 germline variant carriers and their families, including genetic counseling and surveillance for new tumors. Methods We collated germline variant status, tumor diagnoses, and information on BAP1 immunohistochemistry or loss of somatic heterozygosity on 106 published and 75 unpublished BAP1 germline variant-positive families worldwide to better characterize the genotypes and phenotypes associated with the BAP1-TPDS. Tumor spectrum and ages of onset were compared between missense and null variants. All statistical tests were two-sided. Results The 181 families carried 140 unique BAP1 germline variants. The collated data confirmed the core tumor spectrum associated with the BAP1-TPDS and showed that some families carrying missense variants can exhibit this phenotype. A variety of noncore BAP1-TPDS -associated tumors were found in families of variant carriers. Median ages of onset of core tumor types were lower in null than missense variant carriers for all tumors combined (P < .001), mesothelioma (P < .001), cutaneous melanoma (P < .001), and nonmelanoma skin cancer (P < .001). Conclusions This analysis substantially increases the number of pathogenic BAP1 germline variants and refines the phenotype. It highlights the need for a curated registry of germline variant carriers for proper assessment of the clinical phenotype of the BAP1-TPDS and pathogenicity of new variants, thus guiding management of patients and informing areas requiring further research.
Collapse
Affiliation(s)
- Sebastian Walpole
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- University of Queensland, Brisbane, QLD, Australia
| | - Antonia L Pritchard
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- The University of the Highlands and Islands, Inverness, UK
| | - Colleen M Cebulla
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
| | - Robert Pilarski
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Meredith Stautberg
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Frederick H Davidorf
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
| | | | - Odile Cabaret
- Département de Biopathologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Lisa Golmard
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
| | - Dominique Stoppa-Lyonnet
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
- Institut Curie, PSL Research University, INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
- Sorbonne Paris Cité, University Paris-Descartes, Paris, France
| | - Erin Garfield
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Ching-Ni Njauw
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
| | - Mitchell Cheung
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Joni A Turunen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pauliina Repo
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Reetta-Stiina Järvinen
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | | | | | - Cindy Chau
- Department of Ophthalmology, LUMC, Leiden, The Netherlands
| | - Miriam Potrony
- Dermatology Department, Melanoma Unit, Hospital Clinic de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Hildur Helgadottir
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - William Bruno
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Virginia Andreotti
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Giulia Ciccarese
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
| | - Paola Queirolo
- Medical Oncology Unit, Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Department of Surgical and Diagnostic Sciences, Pathology Unit, University of Genoa and Ospedale Policlinico San Martino, Genoa, Italy
| | - Karin Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Jens Folke Kiilgaard
- Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michael R Speicher
- Institute of Human Genetics, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Natasha van Poppelen
- Department of Ophthalmology
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Emine Kilic
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rana’a T Al-Jamal
- Department of Ophthalmology, Ocular Oncology Service, Helsinki University Central Hospital, Helsinki, Finland
| | - Irma Dianzani
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marta Betti
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Carsten Bergmann
- Bioscientia Center for Human Genetics, Ingelheim, Germany
- Department of Medicine IV, Faculty of Medicine, Medical Center—University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Sandro Santagata
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Saleem Taibjee
- Department of Dermatology, Dorset County Hospital NHS Foundation Trust, Dorchester, UK
| | - Jo Burke
- Tasmanian Clinical Genetics Service, Royal Hobart Hospital, TAS, Australia
| | - Nicola Poplawski
- Adult Genetics Unit, Medicine Directorate, Royal Adelaide Hospital, Adelaide, SA, Australia
- University Department of Paediatrics, University of Adelaide, Adelaide, SA, Australia
| | - Sally J O’Shea
- Dermatology Department, Mater Private Hospital Cork, Citygate, Mahon, Cork, Ireland
| | - Julia Newton-Bishop
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Julian Adlard
- Yorkshire Regional Genetics Service, Chapel Allerton Hospital, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
| | - Anne-Marie Lane
- Department of Ophthalmology, Ocular Melanoma Center and Retina Service, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Ivana Kim
- Department of Ophthalmology, Ocular Melanoma Center and Retina Service, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders University and SA Pathology at Flinders Medical Centre, Adelaide, SA, Australia
| | | | - J William Harbour
- Bascom Palmer Eye Institute, Sylvester Comprehensive Cancer Center and Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Michael L Nickerson
- Laboratory of Translational Genomics, National Cancer Institute, Bethesda, MD
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jane M Palmer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Madeleine Howlie
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Judith Symmons
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Hayley Hamilton
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Sunil Warrier
- Queensland Ocular Oncology Service, The Terrace Eye Centre, Brisbane, QLD, Australia
| | - William Glasson
- Queensland Ocular Oncology Service, The Terrace Eye Centre, Brisbane, QLD, Australia
| | - Peter Johansson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Carla Daniela Robles-Espinoza
- Experimental Cancer Genetics, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge, UK
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Santiago de Querétaro, Mexico
| | - Raul Ossio
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, Santiago de Querétaro, Mexico
| | - Annelies de Klein
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Susana Puig
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Raras, Instituto de Salud Carlos III, Barcelona, Spain
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Paola Ghiorzo
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Maartje Nielsen
- Department of Clinical Genetics, LUMC, Leiden, The Netherlands
| | - Tero T Kivelä
- Department of Ophthalmology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hensin Tsao
- Department of Dermatology, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA
- Massachusetts General Hospital Cancer Center, Boston, MA
| | - Joseph R Testa
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA
| | - Pedram Gerami
- Department of Internal Medicine and Medical Specialties and Genetics of Rare Cancers, University of Genoa, Ospedale Policlinico San Martino, Genoa, Italy
- The Robert H. Lurie Cancer Center, Northwestern University, Chicago, IL
| | - Marc-Henri Stern
- Département De Biologie Des Tumeurs, Institut Curie, Paris, France
- Institut Curie, PSL Research University, INSERM U830, DNA Repair and Uveal Melanoma (D.R.U.M.), Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Brigitte Bressac-de Paillerets
- Département de Biopathologie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, PSL, Faculté de Médecine, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Mohamed H Abdel-Rahman
- Department of Ophthalmology and Visual Science, The Ohio State University, Columbus, OH
- Division of Human Genetics, Department of Internal Medicine and Comprehensive Cancer Center, The Ohio State University, Columbus, OH
- Department of Pathology, Menoufiya University, Shebin El-Kom, Egypt
| | | |
Collapse
|
42
|
D'Avella C, Abbosh P, Pal SK, Geynisman DM. Mutations in renal cell carcinoma. Urol Oncol 2018; 38:763-773. [PMID: 30478013 DOI: 10.1016/j.urolonc.2018.10.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is a commonly diagnosed and histologically diverse urologic malignancy. Clear cell RCC (ccRCC) is by far the most common, followed by the papillary and chromophobe subtypes. Sarcomatoid differentiation is a morphologic change that can be seen in all subtypes that typically portends a poor prognosis. In the past, treatment options for RCC were limited to cytokine-based therapy with a high-toxicity profile and low response rate. An increased understanding of the molecular basis of RCC has led to substantial improvement in treatment options in the form of targeted therapy and immunotherapy. A significant early discovery in RCC was frequent inactivation of the Von Hippel Lindau gene in ccRCC, which ultimately led to the development of vascular endothelial growth factor and mammalian target of rapamycin inhibitors. Further genomic sequencing of ccRCC tumors has identified other common mutations including BAP-1, PBRM1, SETD2, and PIK3CA. Many recent studies have explored how these mutations can affect prognosis and response to treatment. Likewise, papillary RCC has also been studied at the molecular level, which has shown a high level of mutations in the MET gene; early clinical data suggest the utility of MET targeted therapy. Finally, regarding the rarer sarcomatoid tumors, mutations in TP53 and NF2 may be important to their development. As we continue to learn more about what drives RCC at the molecular level, treatment options for RCC patients are diversifying.
Collapse
Affiliation(s)
| | - Phillip Abbosh
- Molecular Therapeutics, Fox Chase Cancer Center, Temple Health, Philadelphia, PA; Department of Urology, Einstein Medical Center, Philadelphia, PA
| | - Sumanta K Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Daniel M Geynisman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA.
| |
Collapse
|
43
|
Carbone M, Amelio I, Affar EB, Brugarolas J, Cannon-Albright LA, Cantley LC, Cavenee WK, Chen Z, Croce CM, Andrea AD, Gandara D, Giorgi C, Jia W, Lan Q, Mak TW, Manley JL, Mikoshiba K, Onuchic JN, Pass HI, Pinton P, Prives C, Rothman N, Sebti SM, Turkson J, Wu X, Yang H, Yu H, Melino G. Consensus report of the 8 and 9th Weinman Symposia on Gene x Environment Interaction in carcinogenesis: novel opportunities for precision medicine. Cell Death Differ 2018; 25:1885-1904. [PMID: 30323273 PMCID: PMC6219489 DOI: 10.1038/s41418-018-0213-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/06/2018] [Indexed: 12/13/2022] Open
Abstract
The relative contribution of intrinsic genetic factors and extrinsic environmental ones to cancer aetiology and natural history is a lengthy and debated issue. Gene-environment interactions (G x E) arise when the combined presence of both a germline genetic variant and a known environmental factor modulates the risk of disease more than either one alone. A panel of experts discussed our current understanding of cancer aetiology, known examples of G × E interactions in cancer, and the expanded concept of G × E interactions to include somatic cancer mutations and iatrogenic environmental factors such as anti-cancer treatment. Specific genetic polymorphisms and genetic mutations increase susceptibility to certain carcinogens and may be targeted in the near future for prevention and treatment of cancer patients with novel molecularly based therapies. There was general consensus that a better understanding of the complexity and numerosity of G × E interactions, supported by adequate technological, epidemiological, modelling and statistical resources, will further promote our understanding of cancer and lead to novel preventive and therapeutic approaches.
Collapse
Affiliation(s)
| | | | - El Bachir Affar
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Center, University of Montréal, Montréal, Quebec, H1T 2M4, Canada
| | - James Brugarolas
- Department of Internal Medicine, Hematology-Oncology Division, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, UT, USA
- George E. Wahlen Department of Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medical College, 413 E. 69(th) Street, New York, NY, 10021, USA
| | - Webster K Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhijian Chen
- Department of Molecular Biology and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Alan D' Andrea
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - David Gandara
- Thoracic Oncology, UC Davis, Sacramento, CA, 96817, USA
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Wei Jia
- Hawaii Cancer Center, Honolulu, HI, USA
| | - Qing Lan
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Tak Wah Mak
- The Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada
| | - James L Manley
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Jose N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX, 77005, USA
| | - Harvey I Pass
- Division of General Thoracic Surgery, Department of Cardiothoracic Surgery, NYU Langone Medical Center, New York, NY, USA
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carol Prives
- Department of Biological Sciences, Columbia University, New York, New York, 10027, USA
| | - Nathaniel Rothman
- Occupational & Environmental Epidemiology Branch Division of Cancer Epidemiology & Genetics National Cancer Institute NIH, Bethesda, MD, USA
| | - Said M Sebti
- Drug Discovery Department, Moffitt Cancer Center, and Department of Oncologic Sciences, University of South Florida, Tampa, FL, 33612, USA
| | | | - Xifeng Wu
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Gerry Melino
- MRC Toxicology Unit, Leicester, UK.
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.
| |
Collapse
|
44
|
Pastorino S, Yoshikawa Y, Pass HI, Emi M, Nasu M, Pagano I, Takinishi Y, Yamamoto R, Minaai M, Hashimoto-Tamaoki T, Ohmuraya M, Goto K, Goparaju C, Sarin KY, Tanji M, Bononi A, Napolitano A, Gaudino G, Hesdorffer M, Yang H, Carbone M. A Subset of Mesotheliomas With Improved Survival Occurring in Carriers of BAP1 and Other Germline Mutations. J Clin Oncol 2018; 36:JCO2018790352. [PMID: 30376426 PMCID: PMC7162737 DOI: 10.1200/jco.2018.79.0352] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE We hypothesized that four criteria could help identify malignant mesotheliomas (MMs) most likely linked to germline mutations of BAP1 or of other genes: family history of MM, BAP1-associated cancers, or multiple malignancies; or age younger than 50 years. PATIENTS AND METHODS Over the course of 7 years, 79 patients with MM met the four criteria; 22 of the 79 (28%) reported possible asbestos exposure. They were screened for germline BAP1 mutations by Sanger sequencing and by targeted next-generation sequencing (tNGS) for germline mutations in 55 additional cancer-linked genes. Deleterious mutations detected by tNGS were validated by Sanger sequencing. RESULTS Of the 79 patients, 43 (16 probands and 27 relatives) had deleterious germline BAP1 mutations. The median age at diagnosis was 54 years and median survival was 5 years. Among the remaining 36 patients with no BAP1 mutation, median age at diagnosis was 45 years, median survival was 9 years, and 12 had deleterious mutations of additional genes linked to cancer. When compared with patients with MMs in the SEER cohort, median age at diagnosis (72 years), median survival for all MM stages (8 months), and stage I (11 months) were significantly different from the 79 patients with MM in the current study ( P < .0001). CONCLUSION We provide criteria that help identify a subset of patients with MM who had significantly improved survival. Most of these patients were not aware of asbestos exposure and carried either pathogenic germline mutations of BAP1 or of additional genes linked to cancer, some of which may have targeted-therapy options. These patients and their relatives are susceptible to development of additional cancers; therefore, genetic counseling and cancer screening should be considered.
Collapse
Affiliation(s)
- Sandra Pastorino
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Yoshie Yoshikawa
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Harvey I. Pass
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Mitsuru Emi
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Masaki Nasu
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Ian Pagano
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Yasutaka Takinishi
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Ryuji Yamamoto
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Michael Minaai
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Tomoko Hashimoto-Tamaoki
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Masaki Ohmuraya
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Keisuke Goto
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Chandra Goparaju
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Kavita Y. Sarin
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Mika Tanji
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Angela Bononi
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Andrea Napolitano
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Giovanni Gaudino
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Mary Hesdorffer
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Haining Yang
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| | - Michele Carbone
- Sandra Pastorino, Mitsuru Emi, Masaki Nasu, Ian Pagano, Yasutaka Takinishi, Ryuji Yamamoto, Michael Minaai, Keisuke Goto, Mika Tanji, Angela Bononi, Andrea Napolitano, Giovanni Gaudino, Haining Yang, and Michele Carbone, University of Hawaii Cancer Center, Honolulu, HI; Yoshie Yoshikawa, Mitsuru Emi, Tomoko Hashimoto-Tamaoki, and Masaki Ohmuraya, Hyogo College of Medicine, Hyogo, Japan; Mary Hesdorffer, Mesothelioma Applied Research Foundation, Washington DC; Harvey I. Pass and Chandra Goparaju, New York University Langone Medical Center, New York, NY; Andrea Neopolitano, University Campus Bio-Medico, Rome, Italy; and Kavita Y. Sarin, Stanford University, Stanford, CA
| |
Collapse
|
45
|
Jin S, Wu J, Zhu Y, Gu W, Wan F, Xiao W, Dai B, Zhang H, Shi G, Shen Y, Zhu Y, Ye D. Comprehensive Analysis of BAP1 Somatic Mutation in Clear Cell Renal Cell Carcinoma to Explore Potential Mechanisms in Silico. J Cancer 2018; 9:4108-4116. [PMID: 30519310 PMCID: PMC6277624 DOI: 10.7150/jca.27281] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/24/2018] [Indexed: 12/29/2022] Open
Abstract
Purpose: Aim of this study was to comprehensively analyze BRCA1-associated protein-1 (BAP1) somatic mutation in clear cell renal cell carcinoma (ccRCC) and explore potential therapeutic pathways and molecules. Patients and methods: In this study, we analyzed 445 ccRCC cases from The Cancer Genome Atlas (TCGA). Comprehensive analysis including survival, transcriptome and methylation between BAP1 mutated and wild-type cases was performed using bioinformatics tools in silico. Pathways and molecules related to BAP1 mutation were analyzed using Database for Annotation, Visualization and Integrated Discovery (DAVID) and protein-protein interaction (PPI) network. Results: BAP1 mutated ccRCC patients had a worse overall survival (OS) and disease free survival (DFS) than BAP1 wild-type patients. We found 583 up-regulated and 1216 down-regulated different expressed genes (DEGs) in BAP1 mutated tumors. Up-regulated DEGs were enriched in molecular functions and biological processes like protein binding, protein transport and ubiquitin protein ligase binding. Down-regulated DEGs were enriched in pathways like Rap1 signaling pathway, Notch pathway and altered molecular functions like metal ion binding and ubiquitin-protein transferase activity. Furthermore, CAD, TSPO, CTNNB1 and MAPK3 were top hub genes selected using PPI network analysis. Finally, BAP1 mutation had a strong correlation with CpG island methylator phenotype (CIMP). Conclusion: Our study provides a comprehensive understanding of BAP1 functional somatic mutation in ccRCC patients. Several hub genes like CAD and TSPO may become potential therapeutic targets.
Collapse
Affiliation(s)
- Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weijie Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjun Xiao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yijun Shen
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yiping Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Histomorphologic spectrum of germline-related and sporadic BAP1-inactivated melanocytic tumors. J Am Acad Dermatol 2018; 79:525-534. [DOI: 10.1016/j.jaad.2018.05.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/13/2022]
|
47
|
Imaging for Screening and Surveillance of Patients with Hereditary Forms of Renal Cell Carcinoma. Curr Urol Rep 2018; 19:82. [DOI: 10.1007/s11934-018-0829-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
48
|
Kittaneh M, Berkelhammer C. Detecting germline BAP1 mutations in patients with peritoneal mesothelioma: benefits to patient and family members. J Transl Med 2018; 16:194. [PMID: 30001711 PMCID: PMC6044070 DOI: 10.1186/s12967-018-1559-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/23/2018] [Indexed: 02/01/2023] Open
Abstract
Germline mutations in the BRCA-1 associated tumor protein 1 (BAP1) increase susceptibility to mesothelioma and other cancers. We describe a patient with a family history of peritoneal mesothelioma, who developed malignant peritoneal mesothelioma at age 45 in the absence of known asbestos exposure. These findings lead us to hypothesize that the mesothelioma occurred in the setting of germline a BAP1 mutation. This was confirmed by genetic testing. The subsequent therapeutic choices for the patient and testing of at-risk family members highlight the importance of recognizing this genetic syndrome and screening for individuals at high risk.
Collapse
Affiliation(s)
- Muaiad Kittaneh
- Loyola University, 15300 West Avenue, Orland Park, IL, 60462, USA.
| | | |
Collapse
|
49
|
Hereditary renal cell carcinoma syndromes: diagnosis, surveillance and management. World J Urol 2018; 36:1891-1898. [PMID: 29680948 PMCID: PMC6280834 DOI: 10.1007/s00345-018-2288-5] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 03/31/2018] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Genetic factors have been implicated in the pathogenesis of renal cell carcinoma (RCC), with around 3% of cases having a family history. A greater knowledge of the genetics of inherited RCC has the potential to translate into novel therapeutic targets for sporadic RCC. METHODS A literature review was performed summarising the current knowledge on hereditary RCC diagnosis, surveillance and management. RESULTS Familial RCC is usually inherited in an autosomal dominant manner, although inherited RCC may present without a relevant family history. A number of familial RCC syndromes have been identified. Familial non-syndromic RCC is suspected when ≥ 2 relatives are affected in the absence of syndromic features, although clear diagnostic criteria are lacking. Young age at onset and bilateral/multicentric tumours are recognised characteristics which should prompt molecular genetic analysis. Surveillance in individuals at risk of inherited RCC aims to prevent morbidity and mortality via early detection of tumours. Though screening and management guidelines for some inherited RCC syndromes (e.g. von Hippel-Lindau disease, Birt-Hogg-Dube syndrome, hereditary leiomyomatosis) are well defined for rare cause of inherited RCC (e.g. germline BAP1 mutations), there is limited information regarding the lifetime RCC risks and the most appropriate screening modalities. CONCLUSION Increasing knowledge of the natural history and genetic basis has led to characterisation and tailored management of hereditary RCC syndromes. International data sharing of inherited RCC gene variant information may enable evidence-based improvements in the diagnosis, surveillance protocols and management of these rare conditions.
Collapse
|
50
|
Cardili L, Viana CR, Germano A, Fernandes M, Barcellos D, Landman G. Immunoexpression of BAP1, ROS1, and ALK in Spitzoid Melanocytic Tumors. Int J Surg Pathol 2018; 26:514-520. [PMID: 29623743 DOI: 10.1177/1066896918768089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Spitzoid tumors are a heterogeneous group of melanocytic neoplasms that frequently imposes diagnostic difficulties. Lately, several advances in molecular biology afforded significant discoveries on the pathogenesis of these tumors. BAP1 (BRCA-1 associated protein-1) inactivation and anomalous expression of kinase translocation-related proteins are among the main criteria launched by new classification proposals. Our aim was to systematically assess the immunoexpression of BAP1, ROS1 (receptor tyrosine kinase c-Ros oncogene 1), and ALK (anaplastic lymphoma receptor tyrosine kinase) proteins in an unpublished series of spitzoid tumors. METHODS Retrospective study based on 47 formalin-fixed paraffin-embedded tissue samples from 3 different institutions. BAP1, ROS1, and ALK immunostains were performed in all cases. We included 27 Spitz tumors without significant abnormality, 15 atypical spitzoid tumors, and 5 spitzoid melanomas. RESULTS We observed loss of BAP1 nuclear immunolabeling in 4.3% of evaluable cases (2/46), both of them atypical spitzoid tumors. The proportional frequency of BAP1-inactivated cases among atypical spitzoid tumors was 14.2% (2/14). No immunoexpression of ROS1 or ALK was found. CONCLUSIONS Our study revealed 2 additional BAP1-inactived cases and described its respective frequency. The absence of anomalous expression of translocation-related proteins ALK and ROS1 in this series, composed predominantly of low-grade/low-risk tumors, indicates that translocated spitzoid lesions may not be as prevalent as initially suggested, at least in some populations. Furthermore, our findings encourage additional investigation on unequal occurrence of such immunomarkers among different diagnostic categories of spitzoid neoplasms.
Collapse
Affiliation(s)
- Leonardo Cardili
- 1 Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
- 2 Universidade Federal de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|