51
|
Tomaszewski Z, Littler Y. Unclassified Oncocytic Renal Neoplasm with Rib and Liver Metastases: Metastatic Oncocytoma? Int J Surg Pathol 2022; 30:810-815. [PMID: 35274993 DOI: 10.1177/10668969221084265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Renal tumors with oncocytic or chromophobe-like morphology can be a common source of diagnostic difficulty. In some series, they constitute the largest group of unclassified renal cell carcinomas, a term used for neoplasms that do not fit the current classification of renal tumors. We describe the histological, immunohistochemical, and molecular findings of an eosinophilic renal neoplasm which presented with rib and liver metastases, and provide a review of the literature. The possibility of a renal oncocytoma with metastases was initially considered but excluded on the basis of several morphological and immunohistochemical features. Additionally, the tumor did not correspond with other traditional or newly emerging categories of renal neoplasms. It was therefore regarded as an unclassified oncocytic renal neoplasm which demonstrated evidence of malignant potential due to the presence of multiple metastases.
Collapse
Affiliation(s)
- Zoe Tomaszewski
- Leicester Royal Infirmary, 4490University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Yvonne Littler
- Leicester Royal Infirmary, 4490University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
52
|
Argani P, Mehra R. Renal cell carcinoma associated with tuberous sclerosis complex (TSC)/mammalian target of rapamycin (MTOR) genetic alterations. Mod Pathol 2022; 35:296-297. [PMID: 35046523 DOI: 10.1038/s41379-021-00971-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Pedram Argani
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
53
|
Munari E, Settanni G, Caliò A, Segala D, Lonardi S, Sandrini S, Vacca P, Tumino N, Marconi M, Brunelli M, Gobbo S, Netto GJ, Moretta L, Zamboni G, Martignoni G. TSC loss is a clonal event in eosinophilic solid and cystic renal cell carcinoma: a multiregional tumor sampling study. Mod Pathol 2022; 35:376-385. [PMID: 33990704 DOI: 10.1038/s41379-021-00816-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
Eosinophilic, solid and cystic (ESC) renal cell carcinoma (RCC) is characterized by a solid and cystic architecture with cells showing abundant eosinophilic cytoplasm with hobnail arrangement and a cytokeratin 7-negative/cytokeratin 20-positive immunophenotype. Recent studies have suggested that bi-allelic events affecting TSC genes might play an important role for such tumors. However, only indirect evidence of the clonal origin of TSC mutation has been gathered so far. Therefore, in this paper we aimed to perform multi-regional tumor sampling molecular analysis in four ESC RCC cases that had been completely embedded, three sporadic and one occurring in a patient with tuberous sclerosis complex (TSC). Histologically, the 4 cases showed cystic and solid architecture and cells with abundant eosinophilic cytoplasm with cytoplasmic stippling and round to oval nuclei. Immunohistochemistry showed at least focal expression of cytokeratin 20 in all tissue samples and negative cytokeratin 7, as well as diffuse positivity for S100A1 and at least focal expression of cathepsin K in three out of four cases. The sporadic cases showed the same somatic TSC1 mutations in all tissue samples analyzed, while the TSC-associated case showed the same TSC1 alteration in both normal tissue and all tumor samples analyzed, proving the germline nature of the alteration. In conclusion, our data demonstrate that clonal TSC loss is a key event in ESC RCC and support considering ESC RCC as an entity given its distinct morphologic, immunophenotypical and molecular characteristics.
Collapse
Affiliation(s)
- Enrico Munari
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy.,Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giulio Settanni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Anna Caliò
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Diego Segala
- Pathology Unit, ASST-Spedali Civili, Brescia, Italy
| | - Sara Lonardi
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Silvia Sandrini
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Paola Vacca
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Nicola Tumino
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Marcella Marconi
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy
| | - Matteo Brunelli
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stefano Gobbo
- Pathology Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - George J Netto
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lorenzo Moretta
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Giuseppe Zamboni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Verona, Italy.,Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy. .,Pathology Unit, Pederzoli Hospital, Peschiera del Garda, Verona, Italy.
| |
Collapse
|
54
|
Tuberous Sclerosis Complex (TSC): Renal and Extrarenal Imaging. Acad Radiol 2022; 29:439-449. [PMID: 33487538 DOI: 10.1016/j.acra.2020.12.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/21/2022]
Abstract
Tuberous sclerosis complex is a multiorgan syndrome manifesting with several benign and malignant tumors. Complications arising from renal abnormalities are a leading cause of death in patients with tuberous sclerosis complex. Renal cell carcinoma is relatively uncommon, occurring in 2%-4% of patients with tuberous sclerosis complex syndrome, but nonetheless can significantly contribute to morbidity and mortality. Extrarenal manifestations of tuberous sclerosis complex, including within the chest, abdomen and central nervous system, aid in diagnosis. Pathogenesis and management are also discussed, including the importance of the types of renal masses found in these patients.
Collapse
|
55
|
Kapur P, Gao M, Zhong H, Chintalapati S, Mitui M, Barnes S, Zhou Q, Miyata J, Carrillo D, Malladi V, Rakheja D, Pedrosa I, Xu L, Kinch L, Brugarolas J. Germline and sporadic mTOR pathway mutations in low-grade oncocytic tumor of the kidney. Mod Pathol 2022; 35:333-343. [PMID: 34538873 PMCID: PMC9817016 DOI: 10.1038/s41379-021-00896-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/11/2023]
Abstract
Low-grade oncocytic tumor (LOT) of the kidney is a recently described entity with poorly understood pathogenesis. Using next-generation sequencing (NGS) and complementary approaches, we provide insight into its biology. We describe 22 LOT corresponding to 7 patients presenting with a median age of 75 years (range 63-86 years) and male to female ratio 2:5. All 22 tumors demonstrated prototypical microscopic features. Tumors were well-circumscribed and solid. They were composed of sheets of tumor cells in compact nests. Tumor cells had eosinophilic cytoplasm, round to oval nuclei (without nuclear membrane irregularities), focal subtle perinuclear halos, and occasional binucleation. Sharply delineated edematous stromal islands were often observed. Tumor cells were positive for PAX8, negative for CD117, and exhibited diffuse and strong cytokeratin-7 expression. Six patients presented with pT1 tumors. At a median follow-up of 29 months, four patients were alive without recurrence (three patients had died from unrelated causes). All tumors were originally classified as chromophobe renal cell carcinoma, eosinophilic variant (chRCC-eo). While none of the patients presented with known syndromic features, one patient with multiple bilateral LOTs was subsequently found to have a likely pathogenic germline TSC1 mutation. Somatic, likely activating, mutations in MTOR and RHEB were identified in all other evaluable LOTs. As assessed by phospho-S6 and phospho-4E-BP1, mTOR complex 1 (mTORC1) was activated across all cases but to different extent. MTOR mutant LOT exhibited lower levels of mTORC1 activation, possibly related to mTORC1 dimerization and the preservation of a wild-type MTOR copy (retained chromosome 1). Supporting its distinction from related entities, gene expression analyses showed that LOT clustered separately from classic chRCC, chRCC-eo, and RO. In summary, converging mTORC1 pathway mutations, mTORC1 complex activation, and a distinctive gene expression signature along with characteristic phenotypic features support LOT designation as a distinct entity with both syndromic and non-syndromic cases associated with an indolent course.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ming Gao
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Hua Zhong
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Suneetha Chintalapati
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Midori Mitui
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Spencer Barnes
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Qinbo Zhou
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Jeffrey Miyata
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Deyssy Carrillo
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Venkat Malladi
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Ivan Pedrosa
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, 75390,Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Lin Xu
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Lisa Kinch
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - James Brugarolas
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
56
|
Low-grade oncocytic renal tumor (LOT): mutations in mTOR pathway genes and low expression of FOXI1. Mod Pathol 2022; 35:352-360. [PMID: 34531523 DOI: 10.1038/s41379-021-00906-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 11/09/2022]
Abstract
Low-grade oncocytic renal tumor (LOT) is an emerging provisional entity, described as rare solid renal oncocytic/eosinophilic tumor sharing diffuse CK7 and negative CD117 immunoprofile. The links between LOT and other eosinophilic chromophobe like-renal cell carcinomas (RCC) are currently discussed. We sequenced tumoral DNA with a next generation sequencing panel for kidney cancer and carried out immunohistochemical analyses with CK7, CD117, SDHB, 4EBP1-P, S6K-P, and FOXI1 antibodies in a series of ten cases of LOT (9 females, 1 male; mean age at surgery: 66 years, 42.3 to 83.4) retrospectively diagnosed from a cohort of 272 tumors initially classified as chromophobe RCC (CHRCC). All LOT were single, without known hereditary predisposition, classified stage pT1 (70%), pT2 (20%) or pT3a (10%). Morphological features were similar to previous descriptions and clinical behavior was indolent for the six cases with available follow-up. We identified genetic variations in mTOR pathway related genes in 80% of cases, MTOR (7 cases) or TSC1 (1 case). Expression of FOXI1 was absent in all cases. In 9 LOT, 4EBP1-P and S6K-P were overexpressed, suggesting mTOR pathway activation.Our data highlights the major role of mTOR pathway in tumorigenesis of LOT mostly due to activating MTOR gene variations. Absence of FOXI1 expression is a strong argument to distinguish LOT from eosinophilic CHRCC and to bring them closer to other recently described FOXI1 negative eosinophilic-CHRCC like with MTOR/TSC mutations. Altogether, our data argue to consider LOT as a distinct entity with a favorable clinical outcome. However, in case of metastasis, an accurate diagnosis of LOT would be essential for the patient's management and could allow targeted therapy.
Collapse
|
57
|
Pivovarcikova K, Alaghehbandan R, Vanecek T, Ohashi R, Pitra T, Hes O. TSC/mTOR Pathway Mutation Associated Eosinophilic/Oncocytic Renal Neoplasms: A Heterogeneous Group of Tumors with Distinct Morphology, Immunohistochemical Profile, and Similar Genetic Background. Biomedicines 2022; 10:322. [PMID: 35203531 PMCID: PMC8869370 DOI: 10.3390/biomedicines10020322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
A number of recently described renal tumor entities share an eosinophilic/oncocytic morphology, somewhat solid architectural growth pattern, and tendency to present as low-stage tumors. The vast majority of such tumors follow a non-aggressive clinical behavior. In this review, we discuss the morphological, immunohistochemical, and molecular genetic profiles of the three most recent novel/emerging renal entities associated with TSC/mTOR pathway mutations. These are eosinophilic solid and cystic renal cell carcinoma, eosinophilic vacuolated tumors, and low-grade oncocytic tumors, which belong to a heterogeneous group of renal tumors, demonstrating mostly solid architecture, eosinophilic/oncocytic cytoplasm, and overlapping morphological and immunohistochemical features between renal oncocytoma and chromophobe renal cell carcinoma. All three tumors also share a molecular genetic background with mutations in the mTORC1 pathway (TSC1/TSC2/mTOR/RHEB). Despite the common genetic background, it appears that the tumors with TSC/mTOR mutations represent a diverse group of distinct renal neoplasms.
Collapse
Affiliation(s)
- Kristyna Pivovarcikova
- Department of Pathology, Faculty of Medicine in Pilsen, University Hospital Pilsen, Charles University in Prague, 30460 Pilsen, Czech Republic; (T.V.); (O.H.)
| | - Reza Alaghehbandan
- Department of Pathology, Faculty of Medicine, University of British Columbia, Royal Columbian Hospital, Vancouver, BC V3L 3W7, Canada;
| | - Tomas Vanecek
- Department of Pathology, Faculty of Medicine in Pilsen, University Hospital Pilsen, Charles University in Prague, 30460 Pilsen, Czech Republic; (T.V.); (O.H.)
| | - Riuko Ohashi
- Histopathology Core Facility, Niigata University Faculty of Medicine, 1-757 Asahimachi-dori, Chuo-ku, Niigata 951-8510, Japan;
| | - Tomas Pitra
- Department of Urology, Faculty of Medicine in Pilsen, University Hospital Pilsen, Charles University in Prague, 30599 Pilsen, Czech Republic;
| | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen, University Hospital Pilsen, Charles University in Prague, 30460 Pilsen, Czech Republic; (T.V.); (O.H.)
| |
Collapse
|
58
|
Alaghehbandan R, Przybycin CG, Verkarre V, Mehra R. Chromophobe renal cell carcinoma: Novel molecular insights and clinicopathologic updates. Asian J Urol 2021; 9:1-11. [PMID: 35198391 PMCID: PMC8841285 DOI: 10.1016/j.ajur.2021.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 01/12/2023] Open
|
59
|
Akgul M, Williamson SR. Immunohistochemistry for the diagnosis of renal epithelial neoplasms. Semin Diagn Pathol 2021; 39:1-16. [PMID: 34823973 DOI: 10.1053/j.semdp.2021.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
Despite the increasing number of newly identified renal neoplasms, the diagnosis of renal cell carcinoma (RCC) can usually be reached with careful histologic examination and a limited immunohistochemical (IHC) panel. Clear cell, papillary, chromophobe RCC and oncocytoma account for more than 90% of renal neoplasia in adults, and sophisticated ancillary tools are usually unnecessary. Renal tumors with entity-defining genetic alterations may ultimately require molecular confirmation via cytogenetics or sequencing technologies, such as RCC with TFE3, TFEB, or ALK gene rearrangements, or TFEB amplified RCC. In fumarate hydratase-deficient and succinate dehydrogenase-deficient RCC, highly specific IHC markers can strongly suggest the diagnosis. In the metastatic setting, PAX8 and carbonic anhydrase 9 are among the most helpful markers for confirming RCC and clear cell type, respectively; however, caution should be exercised in the absence of a current or historical renal mass. In diagnostically challenging cases, such as renal eosinophilic tumors with low-grade nuclear features, or infiltrative high-grade tumors, careful examination coupled with a judicious panel of IHC markers usually resolves the diagnosis. This review offers concise algorithms for diagnosis of kidney neoplasia with the latest recognized, provisional, and emerging entities to daily pathology practice.
Collapse
Affiliation(s)
- Mahmut Akgul
- Department of Pathology and Laboratory Medicine, Albany Medical Center, Albany, NY, 12208, USA
| | - Sean R Williamson
- Department of Pathology, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA.
| |
Collapse
|
60
|
Saoud R, Kristof TW, Judge C, Chumbalkar V, Antic T, Eggener S, Modi P. Clinical and pathological features of renal epithelioid angiomyolipoma (PEComa): A single institution series. Urol Oncol 2021; 40:18-24. [PMID: 34815169 DOI: 10.1016/j.urolonc.2021.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/20/2021] [Accepted: 09/20/2021] [Indexed: 10/19/2022]
Abstract
Renal angiomyolipomas are benign tumors of the kidney that belong to the 'PEComa: perivascular epithelioid cell' family. Epithelioid AMLs (eAML) are a rare monotypic subtype with malignant potential, that can occur sporadically or be associated with tuberous sclerosis (TSC). Due to their epithelioid nature, eAMLs can closely resemble high-grade renal cell carcinoma (RCC), which may result in misdiagnosis. Multiple clinicopathologic parameters are predictive of worse outcomes for patients with eAML. Those can be used to stratify patients into groups with low, intermediate and high risk for disease progression. A high index of suspicion and a thorough immunohistochemical study are required to correctly diagnose eAML. Radiographically, eAMLs are also a diagnostic challenge as they share features with RCC on CT and MR imaging. Due to this close mimicry, the true incidence of eAML is thought to be much higher than 200 cases as reported in the literature. We report a series of four patients diagnosed with eAML and compare their clinical courses. We also report on the successful treatment of a patient with pulmonary metastasis from eAML using the mTOR inhibitor, everolimus. By identifying eAML and recognizing its high-risk features, it is possible mTOR inhibitors may have a meaningful role in the adjuvant treatment of these patients.
Collapse
Affiliation(s)
- Ragheed Saoud
- Section of Urology, Department of Surgery, The University of Chicago Medicine, Chicago, IIllinois.
| | - Tanya W Kristof
- Section of Urology, Department of Surgery, The University of Chicago Medicine, Chicago, IIllinois
| | - Clark Judge
- Section of Urology, Department of Surgery, The University of Chicago Medicine, Chicago, IIllinois
| | - Vaibhav Chumbalkar
- Department of Pathology, The University of Chicago Medicine, Chicago, IIllinois
| | - Tatjana Antic
- Department of Pathology, The University of Chicago Medicine, Chicago, IIllinois
| | - Scott Eggener
- Section of Urology, Department of Surgery, The University of Chicago Medicine, Chicago, IIllinois
| | - Parth Modi
- Section of Urology, Department of Surgery, The University of Chicago Medicine, Chicago, IIllinois
| |
Collapse
|
61
|
Kwon HJ, Jang MH. Chromophobe renal cell carcinoma-like thyroid carcinoma: possible misdiagnosis as metastatic renal cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:1095-1101. [PMID: 34900078 PMCID: PMC8661068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/15/2021] [Indexed: 06/14/2023]
Abstract
To date, multiple thyroid cancer variants have been reported. Herein, we report a rare case of chromophobe renal cell carcinoma-like thyroid carcinoma (CRETHCA) in a 60-year-old woman, for which the morphologic findings resembled those of chromophobe renal cell carcinoma (ChRCC). ChRCC of the kidney is characterized by large polygonal tumor cells with distinct cell borders, perinuclear clearing, multiple binucleate cells, and strongly positive immunostaining for paired box gene 8 (PAX8) and carbonic anhydrase IX (CA IX). In our case, the thyroid gland tumor was incidentally detected by routine medical screening without sufficient medical information; it showed similar histology and immunohistochemical features to ChRCC and was initially misdiagnosed as metastatic ChRCC. Additional tests, including kidney computed tomography and positron emission tomography, revealed no abnormalities in the patient's kidney; therefore, we diagnosed the tumor as CRETHCA. Focal weak staining for thyroid transcription factor 1 (TTF-1) was the only supporting evidence that it was a primary thyroid neoplasm. To the best of our knowledge, this is the second report of CRETHCA in literature. This novel variant is very difficult to distinguish from metastatic ChRCC and can be a diagnostic challenge for pathologists. Further studies of similar cases should be done to define this new entity.
Collapse
Affiliation(s)
- Hee Jung Kwon
- Department of Pathology, Yeungnam University College of Medicine Daegu, South Korea
| | - Min Hye Jang
- Department of Pathology, Yeungnam University College of Medicine Daegu, South Korea
| |
Collapse
|
62
|
[Renal carcinoma with leiomyomatous stroma and osseous metaplasia from a patient diagnosed with a tuberous sclerosis complex]. Ann Pathol 2021; 41:557-560. [PMID: 34629215 DOI: 10.1016/j.annpat.2021.06.005] [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: 12/17/2018] [Revised: 01/22/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022]
Abstract
Renal cell carcinoma with leiomyomatous stroma is a rare and poorly described histopathological entity. Here we report a unique case with osseous metaplasia, in a 31-year-old man recently diagnosed with a tuberous sclerosis complex (TSC2 gene mutation). Partial nephrectomy was performed. Histologically, the epithelial component was made up of papillary and alveolar structures with clear to eosinophilic cytoplasm, and basally located nuclei. The cells are surrounded by an abundant smooth muscle stroma with focally osseous metaplasia. The tumor was positive for carbonic anhydrase IX, cytokeratin 7, cytokeratin 20, and CD10, and negative for TFE3. This emerging entity is highly correlated to tuberous sclerosis complex, which justifies a screening for the syndrome when this diagnosis is made.
Collapse
|
63
|
Henske EP, Cornejo KM, Wu CL. Renal Cell Carcinoma in Tuberous Sclerosis Complex. Genes (Basel) 2021; 12:1585. [PMID: 34680979 PMCID: PMC8535193 DOI: 10.3390/genes12101585] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 12/17/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant disorder in which renal manifestations are prominent. There are three major renal lesions in TSC: angiomyolipomas, cysts, and renal cell carcinoma (RCC). Major recent advances have revolutionized our understanding of TSC-associated RCC, including two series that together include more than 100 TSC-RCC cases, demonstrating a mean age at onset of about 36 years, tumors in children as young as 7, and a striking 2:1 female predominance. These series also provide the first detailed understanding of the pathologic features of these distinctive tumors, which include chromophobe-like features and eosinophilia, with some of the tumors unclassified. This pathologic heterogeneity is distinctive and reminiscent of the pathologic heterogeneity in Birt-Hogg-Dube-associated RCC, which also includes chromophobe-like tumors. Additional advances include the identification of sporadic counterpart tumors that carry somatic TSC1/TSC2/mTOR mutations. These include unclassified eosinophilic tumors, eosinophilic solid cystic RCC (ESC-RCC), and RCC with leiomyomatous stroma (RCCLMS). A variety of epithelial renal neoplasms have been identified both in patients with tuberous sclerosis complex (TSC) and in the nonsyndromic setting associated with somatic mutations in the TSC1 and TSC2 genes. Interestingly, whether tumors are related to a germline or somatic TSC1/2 mutation, these tumors often display similar morphologic and immunophenotypic features. Finally, recent work has identified molecular links between TSC and BHD-associated tumors, involving the TFEB/TFE3 transcription factors.
Collapse
Affiliation(s)
- Elizabeth P. Henske
- Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kristine M. Cornejo
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (K.M.C.); (C.-L.W.)
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (K.M.C.); (C.-L.W.)
| |
Collapse
|
64
|
Tran J, Ornstein MC. Clinical Review on the Management of Metastatic Renal Cell Carcinoma. JCO Oncol Pract 2021; 18:187-196. [PMID: 34529499 DOI: 10.1200/op.21.00419] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinomas vary considerably in their tumor biology and disease course, which is reflected in the range of treatment paradigms in localized and metastatic renal cell carcinoma (mRCC). Active surveillance remains an important component of all renal cell carcinoma management. In mRCC, the rapid evolution from cytokine-based therapy to targeted therapy to immunotherapy with checkpoint blockade has revolutionized the field and drastically altered treatment outcomes. More recently, combination therapies have become a standard of care for most patients with mRCC. In this review, we highlight recent critical data that led to changes in treatment paradigms and provide a practical framework for the management of patients with mRCC.
Collapse
Affiliation(s)
- Jennifer Tran
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH
| | - Moshe C Ornstein
- Department of Hematology & Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| |
Collapse
|
65
|
Kirby E, Elbashier S, Sweeney C, Twomey E, Gorman K, Riordan M, Awan A. Unexpected diagnosis in a child with hemolytic uremic syndrome: Answers. Pediatr Nephrol 2021; 36:2711-2714. [PMID: 33730282 DOI: 10.1007/s00467-021-05005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/05/2021] [Indexed: 11/26/2022]
Affiliation(s)
- Eimear Kirby
- School of Medicine, Trinity College Dublin, University of Dublin, College Green, Dublin 2, Ireland.
| | - Shiraz Elbashier
- Department of Pediatric Nephrology, Temple Street Children's Hospital, Dublin, Ireland
| | - Clodagh Sweeney
- Department of Pediatric Nephrology, Temple Street Children's Hospital, Dublin, Ireland
| | - Eilish Twomey
- Department of Pediatric Radiology, Temple Street Children's Hospital, Dublin, Ireland
| | - Kathleen Gorman
- Department of Pediatric Neurology, Temple Street Children's Hospital, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| | - Michael Riordan
- Department of Pediatric Nephrology, Temple Street Children's Hospital, Dublin, Ireland
- Faculty of Medicine and Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Atif Awan
- Department of Pediatric Nephrology, Temple Street Children's Hospital, Dublin, Ireland
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
- Faculty of Medicine and Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
66
|
Chakraborty S, Balan M, Sabarwal A, Choueiri TK, Pal S. Metabolic reprogramming in renal cancer: Events of a metabolic disease. Biochim Biophys Acta Rev Cancer 2021; 1876:188559. [PMID: 33965513 PMCID: PMC8349779 DOI: 10.1016/j.bbcan.2021.188559] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/21/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022]
Abstract
Recent studies have established that tumors can reprogram the pathways involved in nutrient uptake and metabolism to withstand the altered biosynthetic, bioenergetics and redox requirements of cancer cells. This phenomenon is called metabolic reprogramming, which is promoted by the loss of tumor suppressor genes and activation of oncogenes. Because of alterations and perturbations in multiple metabolic pathways, renal cell carcinoma (RCC) is sometimes termed as a "metabolic disease". The majority of metabolic reprogramming in renal cancer is caused by the inactivation of von Hippel-Lindau (VHL) gene and activation of the Ras-PI3K-AKT-mTOR pathway. Hypoxia-inducible factor (HIF) and Myc are other important players in the metabolic reprogramming of RCC. All types of RCCs are associated with reprogramming of glucose and fatty acid metabolism and the tricarboxylic acid (TCA) cycle. Metabolism of glutamine, tryptophan and arginine is also reprogrammed in renal cancer to favor tumor growth and oncogenesis. Together, understanding these modifications or reprogramming of the metabolic pathways in detail offer ample opportunities for the development of new therapeutic targets and strategies, discovery of biomarkers and identification of effective tumor detection methods.
Collapse
Affiliation(s)
- Samik Chakraborty
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Murugabaskar Balan
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Akash Sabarwal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Toni K Choueiri
- Dana Farber Cancer Institute, Boston, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America
| | - Soumitro Pal
- Division of Nephrology, Boston Children's Hospital, MA 02115, United States of America; Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
67
|
TSC2 regulates lysosome biogenesis via a non-canonical RAGC and TFEB-dependent mechanism. Nat Commun 2021; 12:4245. [PMID: 34253722 PMCID: PMC8275687 DOI: 10.1038/s41467-021-24499-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
Tuberous Sclerosis Complex (TSC) is caused by TSC1 or TSC2 mutations, resulting in hyperactivation of the mechanistic target of rapamycin complex 1 (mTORC1). Transcription factor EB (TFEB), a master regulator of lysosome biogenesis, is negatively regulated by mTORC1 through a RAG GTPase-dependent phosphorylation. Here we show that lysosomal biogenesis is increased in TSC-associated renal tumors, pulmonary lymphangioleiomyomatosis, kidneys from Tsc2+/- mice, and TSC1/2-deficient cells via a TFEB-dependent mechanism. Interestingly, in TSC1/2-deficient cells, TFEB is hypo-phosphorylated at mTORC1-dependent sites, indicating that mTORC1 is unable to phosphorylate TFEB in the absence of the TSC1/2 complex. Importantly, overexpression of folliculin (FLCN), a GTPase activating protein for RAGC, increases TFEB phosphorylation at the mTORC1 sites in TSC2-deficient cells. Overexpression of constitutively active RAGC is sufficient to relocalize TFEB to the cytoplasm. These findings establish the TSC proteins as critical regulators of lysosomal biogenesis via TFEB and RAGC and identify TFEB as a driver of the proliferation of TSC2-deficient cells.
Collapse
|
68
|
Sauter M, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D'Amato L, d'Augères GB, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Jansen A, Kingswood JC. Rare manifestations and malignancies in tuberous sclerosis complex: findings from the TuberOus SClerosis registry to increAse disease awareness (TOSCA). Orphanet J Rare Dis 2021; 16:301. [PMID: 34229737 PMCID: PMC8259106 DOI: 10.1186/s13023-021-01917-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tuberous sclerosis complex (TSC) is a rare multisystem autosomal dominant disorder caused by pathogenic variants in either the TSC1 or TSC2 gene. Common manifestations of TSC have been grouped into major and minor clinical diagnostic criteria and assessed in clinical routine workup. However, case studies point towards the existence of rare disease manifestations and to the potential association of TSC with malignant tumors. In this study we sought to characterize rare manifestations and malignancies using a large cohort of patients. METHODS TuberOus SClerosis registry to increAse disease awareness (TOSCA) is a multicenter, international disease registry collecting clinical manifestations and characteristics of patients with TSC, both retrospectively and prospectively. We report rates and characteristics of rare manifestations and malignancies in patients with TSC who had enrolled in the TOSCA registry. We also examined these manifestations by age, sex, and genotype (TSC1 or TSC2). RESULTS Overall, 2211 patients with TSC were enrolled in the study. Rare manifestations were reported in 382 (17.3%) study participants and malignancies in 65 (2.9%). Of these rare manifestations, the most frequent were bone sclerotic foci (39.5%), scoliosis (23%), thyroid adenoma (5.5%), adrenal angiomyolipoma (4.5%), hemihypertrophy and pancreatic neuroendocrine tumors (pNET; both 3.1%). These rare manifestations were more commonly observed in adults than children (66.2% vs. 22.7%), in females versus males (58.4% vs. 41.6%; except for scoliosis: 48.9% vs. 51.1%), and in those with TSC2 versus TSC1 (67.0% vs. 21.1%; except for thyroid adenoma: 42.9% vs. 57.1%). In the 65 individuals with reported malignancies, the most common were renal cell carcinoma (47.7%), followed by breast (10.8%) and thyroid cancer (9.2%). Although malignancies were more common in adult patients, 26.1% were reported in children and 63.1% in individuals < 40 years. TSC1 mutations were over-represented in individuals with malignancies compared to the overall TOSCA cohort (32.1% vs. 18.5%). CONCLUSION Rare manifestations were observed in a significant proportion of individuals with TSC. We recommend further examination of rare manifestations in TSC. Collectively, malignancies were infrequent findings in our cohort. However, compared to the general population, malignant tumors occurred earlier in age and some tumor types were more common.
Collapse
Affiliation(s)
- Matthias Sauter
- Klinikum Kempten, Klinikverbund Allgäu, Robert-Weixler-Str. 50, 87439, Kempten, Germany.
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | | | - Tom Carter
- TSA Tuberous Sclerosis Association, Nottingham, UK
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | | | - Maria Dahlin
- Karolinska University Hospital, Stockholm, Sweden
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | | | - Martha Feucht
- Universitätsklinik Für Kinder-Und Jugendheilkunde (Affiliated Partner of the ERN EpiCARE), Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,In Den Birken, European Tuberous Sclerosis Complex Association, Dattein, Germany
| | | | - Sergiusz Jozwiak
- Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.P.A, Origgio, Italy.,Institute of Biomedicine (IBIOMED), University of León, León, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Imagine Institute, Paris Descartes University, Paris, France
| | | | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
| | | | - Seema Shah
- Novartis Healthcare Pvt. Ltd, Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, Aoi-ku, UrushiyamaShizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint Etienne, France
| | | | | | - Anna Jansen
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel VUB, Brussels, Belgium
| | - J Chris Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St Georges University of London, London, UK
| | | |
Collapse
|
69
|
Peired AJ, Lazzeri E, Guzzi F, Anders HJ, Romagnani P. From kidney injury to kidney cancer. Kidney Int 2021; 100:55-66. [PMID: 33794229 DOI: 10.1016/j.kint.2021.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/04/2021] [Accepted: 02/17/2021] [Indexed: 02/07/2023]
Abstract
Epidemiologic studies document strong associations between acute or chronic kidney injury and kidney tumors. However, whether these associations are linked by causation, and in which direction, is unclear. Accumulating data from basic and clinical research now shed light on this issue and prompt us to propose a new pathophysiological concept with immanent implications in the management of patients with kidney disease and patients with kidney tumors. As a central paradigm, this review proposes the mechanisms of kidney damage and repair that are active during acute kidney injury but also during persistent injuries in chronic kidney disease as triggers of DNA damage, promoting the expansion of (pre-)malignant cell clones. As renal progenitors have been identified by different studies as the cell of origin for several benign and malignant kidney tumors, we discuss how the different types of kidney tumors relate to renal progenitors at specific sites of injury and to germline or somatic mutations in distinct signaling pathways. We explain how known risk factors for kidney cancer rather represent risk factors for kidney injury as an upstream cause of cancer. Finally, we propose a new role for nephrologists in kidney cancer (i.e., the primary and secondary prevention and treatment of kidney injury to reduce incidence, prevalence, and recurrence of kidney cancer).
Collapse
Affiliation(s)
- Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies, University of Florence, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies, University of Florence, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Ludwig Maximilian University Klinikum, Munich, Germany
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies, University of Florence, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy; Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy.
| |
Collapse
|
70
|
Baniak N, Barletta JA, Hirsch MS. Key Renal Neoplasms With a Female Predominance. Adv Anat Pathol 2021; 28:228-250. [PMID: 34009777 DOI: 10.1097/pap.0000000000000301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Renal neoplasms largely favor male patients; however, there is a growing list of tumors that are more frequently diagnosed in females. These tumors include metanephric adenoma, mixed epithelial and stromal tumor, juxtaglomerular cell tumor, mucinous tubular and spindle cell carcinoma, Xp11.2 (TFE3) translocation-associated renal cell carcinoma, and tuberous sclerosis complex (somatic or germline) associated renal neoplasms. The latter category is a heterogenous group with entities still being delineated. Eosinophilic solid and cystic renal cell carcinoma is the best-described entity, whereas, eosinophilic vacuolated tumor is a proposed entity, and the remaining tumors are currently grouped together under the umbrella of tuberous sclerosis complex/mammalian target of rapamycin-related renal neoplasms. The entities described in this review are often diagnostic considerations when evaluating renal mass tissue on biopsy or resection. For example, Xp11.2 translocation renal cell carcinoma is in the differential when a tumor has clear cell cytology and papillary architecture and occurs in a young or middle-aged patient. In contrast, tuberous sclerosis complex-related neoplasms often enter the differential for tumors with eosinophilic cytology. This review provides an overview of the clinical, gross, microscopic, immunohistochemical, genetic, and molecular alterations in key renal neoplasms occurring more commonly in females; differential diagnoses are also discussed regardless of sex predilection.
Collapse
Affiliation(s)
- Nicholas Baniak
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
71
|
Meng F, Zhang L, Zhang M, Ye K, Guo W, Liu Y, Yang W, Zhai Z, Wang H, Xiao J, Dai H. Down-regulation of BCL2L13 renders poor prognosis in clear cell and papillary renal cell carcinoma. Cancer Cell Int 2021; 21:332. [PMID: 34193180 PMCID: PMC8247248 DOI: 10.1186/s12935-021-02039-y] [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: 03/04/2021] [Accepted: 06/22/2021] [Indexed: 11/15/2022] Open
Abstract
Background BCL2L13 belongs to the BCL2 super family, with its protein product exhibits capacity of apoptosis-mediating in diversified cell lines. Previous studies have shown that BCL2L13 has functional consequence in several tumor types, including ALL and GBM, however, its function in kidney cancer remains as yet unclearly. Methods Multiple web-based portals were employed to analyze the effect of BCL2L13 in kidney cancer using the data from TCGA database. Functional enrichment analysis and hubs of BCL2L13 co-expressed genes in clear cell renal cell carcinoma (ccRCC) and papillary renal cell carcinoma (pRCC) were carried out on Cytoscape. Evaluation of BCL2L13 protein level was accomplished through immunohistochemistry on paraffin embedded renal cancer tissue sections. Western blotting and flow cytometry were implemented to further analyze the pro-apoptotic function of BCL2L13 in ccRCC cell line 786-0. Results BCL2L13 expression is significantly decreased in ccRCC and pRCC patients, however, mutations and copy number alterations are rarely observed. The poor prognosis of ccRCC that derived from down-regulated BCL2L13 is independent of patients’ gender or tumor grade. Furthermore, BCL2L13 only weakly correlates with the genes that mutated in kidney cancer or the genes that associated with inherited kidney cancer predisposing syndrome, while actively correlates with SLC25A4. As a downstream effector of BCL2L13 in its pro-apoptotic pathway, SLC25A4 is found as one of the hub genes that involved in the physiological function of BCL2L13 in kidney cancer tissues. Conclusions Down-regulation of BCL2L13 renders poor prognosis in ccRCC and pRCC. This disadvantageous factor is independent of any well-known kidney cancer related genes, so BCL2L13 can be used as an effective indicator for prognostic evaluation of renal cell carcinoma. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02039-y.
Collapse
Affiliation(s)
- Fei Meng
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,University of Science and Technology of China, Hefei, 230026, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Luojin Zhang
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Mingjun Zhang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Kaiqin Ye
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wei Guo
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,University of Science and Technology of China, Hefei, 230026, China
| | - Yu Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,University of Science and Technology of China, Hefei, 230026, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wulin Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Zhimin Zhai
- Department of Hematology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jun Xiao
- Department of Urology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Haiming Dai
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, 350 Shushanhu Road, Hefei, 230031, Anhui, China. .,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China.
| |
Collapse
|
72
|
Lerma LA, Schade GR, Tretiakova MS. Co-existence of ESC-RCC, EVT, and LOT as synchronous and metachronous tumors in six patients with multifocal neoplasia but without clinical features of tuberous sclerosis complex. Hum Pathol 2021; 116:1-11. [PMID: 34153307 DOI: 10.1016/j.humpath.2021.06.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/23/2021] [Accepted: 06/07/2021] [Indexed: 12/19/2022]
Abstract
Renal cell tumors with oncocytic phenotypes represent a daily challenge, with several novel, emerging, and provisional entities enriching the diagnostic repertoire. Eosinophilic solid and cystic renal cell carcinoma (ESC-RCC), low-grade oncocytic tumor (LOT), and eosinophilic vacuolated tumor (EVT) have been recognized as unique entities, although their distinctive nature remains controversial. Although most of these tumors are sporadic, rare reports of similar tumors in tuberous sclerosis complex (TSC) have been published. We describe multifocal, often bilateral, tumors in six patients without personal or family history of syndromic diseases. More than 60 tumors in various combinations were identified in 10 nephrectomies and one biopsy encompassing ESC-RCC (n = 6), LOT (n = 14), EVT (n = 1), clear cell RCC with fibromyomatous stroma (n = 12), clear cell RCC (n = 2), angiomyolipomas (AMLs; n > 20), unclassified renal cell tumors (n = 2), papillary adenomas (n = 4), and renomedullary interstitial cell tumor (n = 1). TSC1 germline pathogenic mutations were confirmed in two patients. A tumor without germline testing in a third patient revealed TSC1 biallelic inactivation. Two additional patients had molecular testing, which excluded common renal mutations and syndromes. We provide the first evidence of co-existence in the same organ and unequivocal relatedness of ESC-RCC, EVT, and LOT. End-stage renal disease was present in three of six patients with precursor lesions to all above tumors within adjacent renal parenchyma. In conclusion, identification of multifocal tumors with TSC-like morphology, especially in association with AMLs, could be the first manifestation of clinically silent TSC guiding clinical recommendations for further genetic testing and/or treatment recommendations.
Collapse
Affiliation(s)
- L Angelica Lerma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.
| | - George R Schade
- Department of Urology, University of Washington, Seattle, WA, 98195, USA
| | - Maria S Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
73
|
Marchetti A, Rosellini M, Mollica V, Rizzo A, Tassinari E, Nuvola G, Cimadamore A, Santoni M, Fiorentino M, Montironi R, Massari F. The Molecular Characteristics of Non-Clear Cell Renal Cell Carcinoma: What's the Story Morning Glory? Int J Mol Sci 2021; 22:6237. [PMID: 34207825 PMCID: PMC8226484 DOI: 10.3390/ijms22126237] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 01/05/2023] Open
Abstract
Non-clear cell renal cell carcinomas are a miscellaneous group of tumors that include different histological subtypes, each one characterized by peculiarity in terms of genetic alteration, clinical behavior, prognosis, and treatment response. Because of their low incidence and poor enrollment in clinical trials, alongside their heterogeneity, additional efforts are required to better unveil the pathogenetic mechanisms and, consequently, to improve the treatment algorithm. Nowadays, tyrosine kinase inhibitors, mTOR and MET inhibitors, and even cisplatin-based chemotherapy and immunotherapy are potential weapons that are still under evaluation in this setting. Various biomarkers have been evaluated for detecting progression and monitoring renal cell carcinoma, but more studies are necessary to improve this field. In this review, we provide an overview on the molecular characteristics of this group of tumors and the recently published trials, giving an insight into what might become the future therapeutic standard in this complex world of non-clear cell kidney cancers.
Collapse
Affiliation(s)
- Andrea Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Matteo Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Elisa Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Giacomo Nuvola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Michelangelo Fiorentino
- Department of Specialistic Diagnostic and Experimental Medicine, University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, Polytechnic University of the Marche Region, United Hospitals, 60126 Ancona, Italy; (A.C.); (R.M.)
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni—15, 40138 Bologna, Italy; (A.M.); (M.R.); (V.M.); (A.R.); (E.T.); (G.N.)
| |
Collapse
|
74
|
Renal tumors in tuberous sclerosis complex. Pediatr Nephrol 2021; 36:1427-1438. [PMID: 33006051 DOI: 10.1007/s00467-020-04775-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/11/2020] [Accepted: 09/10/2020] [Indexed: 01/19/2023]
Abstract
Tuberous sclerosis complex (TSC) is a multisystem hereditary disorder characterized by the growth of benign tumors (hamartomas) in multiple organs, including the kidneys. Renal angiomyolipomas (AML) are a major diagnostic feature of TSC and are present in the majority of patients by adulthood. However, AML are usually asymptomatic during childhood when neurological and developmental manifestations are the main source of morbidity. Kidney manifestations of TSC have historically been the main cause of morbidity and mortality of adults with TSC. The recognition that the complications of TSC are caused by dysregulation of the mammalian target of rapamycin (mTOR) pathway has led to an enormous progress in the management of patients with TSC in the last two decades, the establishment of diagnostic guidelines, and trials which have shown the therapeutic benefit of mTOR inhibitors. Kidney surveillance of children with TSC now provides the opportunity for timely interventions to reduce the impact of TSC in adulthood. In this review, we discuss the current management of kidney tumors associated with TSC, including the diagnosis, surveillance, and treatment options for these lesions. We also present outcome data from international registries demonstrating the effectiveness of the current management strategies. With clear management guidelines and efficient treatment of kidney tumors, we envisage that the long-term outcomes of patients with TSC will further improve in the future.
Collapse
|
75
|
Trpkov K, Williamson SR, Gill AJ, Adeniran AJ, Agaimy A, Alaghehbandan R, Amin MB, Argani P, Chen YB, Cheng L, Epstein JI, Cheville JC, Comperat E, da Cunha IW, Gordetsky JB, Gupta S, He H, Hirsch MS, Humphrey PA, Kapur P, Kojima F, Lopez JI, Maclean F, Magi-Galluzzi C, McKenney JK, Mehra R, Menon S, Netto GJ, Przybycin CG, Rao P, Rao Q, Reuter VE, Saleeb RM, Shah RB, Smith SC, Tickoo S, Tretiakova MS, True L, Verkarre V, Wobker SE, Zhou M, Hes O. Novel, emerging and provisional renal entities: The Genitourinary Pathology Society (GUPS) update on renal neoplasia. Mod Pathol 2021; 34:1167-1184. [PMID: 33526874 DOI: 10.1038/s41379-021-00737-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
The Genitourinary Pathology Society (GUPS) undertook a critical review of the recent advances in renal neoplasia, particularly focusing on the newly accumulated evidence post-2016 World Health Organization (WHO) classification. In the era of evolving histo-molecular classification of renal neoplasia, morphology is still key. However, entities (or groups of entities) are increasingly characterized by specific molecular features, often associated either with recognizable, specific morphologies or constellations of morphologies and corresponding immunohistochemical profiles. The correct diagnosis has clinical implications leading to better prognosis, potential clinical management with targeted therapies, may identify hereditary or syndromic associations, which may necessitate appropriate genetic testing. We hope that this undertaking will further facilitate the identification of these entities in practice. We also hope that this update will bring more clarity regarding the evolving classification of renal neoplasia and will further reduce the category of "unclassifiable renal carcinomas/tumors". We propose three categories of novel entities: (1) "Novel entity", validated by multiple independent studies; (2) "Emerging entity", good compelling data available from at least two or more independent studies, but additional validation is needed; and (3) "Provisional entity", limited data available from one or two studies, with more work required to validate them. For some entities initially described using different names, we propose new terminologies, to facilitate their recognition and to avoid further diagnostic dilemmas. Following these criteria, we propose as novel entities: eosinophilic solid and cystic renal cell carcinoma (ESC RCC), renal cell carcinoma with fibromyomatous stroma (RCC FMS) (formerly RCC with leiomyomatous or smooth muscle stroma), and anaplastic lymphoma kinase rearrangement-associated renal cell carcinoma (ALK-RCC). Emerging entities include: eosinophilic vacuolated tumor (EVT) and thyroid-like follicular renal cell carcinoma (TLFRCC). Finally, as provisional entities, we propose low-grade oncocytic tumor (LOT), atrophic kidney-like lesion (AKLL), and biphasic hyalinizing psammomatous renal cell carcinoma (BHP RCC).
Collapse
Affiliation(s)
- Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Sean R Williamson
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anthony J Gill
- Sydney Medical School, University of Sydney; Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research; NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, Sydney, NSW, Australia
| | | | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Erlangen, Germany
| | - Reza Alaghehbandan
- Department of Pathology, Faculty of Medicine, University of British Columbia, Royal Columbian Hospital, Vancouver, BC, Canada
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science, Memphis, TN, USA
| | - Pedram Argani
- Departments of Pathology and Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jonathan I Epstein
- Departments of Pathology, Urology and Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | - Eva Comperat
- Department of Pathology, Hôpital Tenon, Sorbonne University, Paris, France
| | | | - Jennifer B Gordetsky
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sounak Gupta
- Department of Pathology, Mayo Clinic, Rochester, MN, USA
| | - Huiying He
- Department of Pathology, Health Science Center, Peking University, Beijing, China
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter A Humphrey
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Payal Kapur
- Departments of Pathology, Urology, Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fumiyoshi Kojima
- Department of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Jose I Lopez
- Department of Pathology, Cruces University Hospital, Biocruces-Bizkaia Institute, Bizkaia, Spain
| | - Fiona Maclean
- Department of Clinical Medicine, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, Australia.,Anatomical Pathology, Douglass Hanly Moir Pathology, Sydney, Australia
| | | | - Jesse K McKenney
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rohit Mehra
- Department of Pathology and Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Santosh Menon
- Department of Surgical Pathology, Tata Memorial Hospital, Parel, and Homi Bhabha National Institute, Mumbai, India
| | - George J Netto
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christopher G Przybycin
- Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Priya Rao
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Victor E Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rola M Saleeb
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven C Smith
- Departments of Pathology and Urology, VCU School of Medicine, Richmond, VA, USA
| | - Satish Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maria S Tretiakova
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Lawrence True
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| | - Virginie Verkarre
- Department of Pathology, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris; Université de Paris, PARCC, INSERM, Equipe Labellisée par la Ligue contre le Cancer, F-75015, Paris, France
| | - Sara E Wobker
- Departments of Pathology and Laboratory Medicine and Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ming Zhou
- Department of Pathology, Tufts Medical Center, Boston, MA, USA
| | - Ondrej Hes
- Department of Pathology, Charles University in Prague, Faculty of Medicine and University Hospital in Plzen, Plzen, Czech Republic
| |
Collapse
|
76
|
Gupta S, Jimenez RE, Herrera-Hernandez L, Lohse CM, Thompson RH, Boorjian SA, Leibovich BC, Cheville JC. Renal Neoplasia in Tuberous Sclerosis: A Study of 41 Patients. Mayo Clin Proc 2021; 96:1470-1489. [PMID: 33526281 DOI: 10.1016/j.mayocp.2020.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To study the clinical features and identify unique renal neoplasia subtypes and their prognostic implications in individuals with tuberous sclerosis complex (TSC). PATIENTS AND METHODS The Mayo Clinic nephrectomy registry included 37 patients with TSC diagnosed between 1970 and 2018. Four additional patients were identified from the pathology consultation and autopsy files. All available renal tumors were further characterized using immunohistochemistry and fluorescence in situ hybridization. Clinicopathologic features and follow-up were obtained from the medical record. The American Association for Cancer Research Project GENIE registry was accessed using cBioPortal for molecular profiling of angiomyolipoma (AML). RESULTS A total of 276 renal tumors from 41 patients were analyzed. Renal tumors were classified into 9 distinct morphological subtypes, with AML predominating (238 [86%]). Interestingly, all these tumors acted in a benign fashion except one renal cell carcinoma with clear cells and fibromyomatous stroma and one epithelioid AML that metastasized. Molecular profiling studies revealed that epithelioid AMLs were enriched for alterations of TP53, RB1, and ATRX. Eight patients died of direct complications of TSC, including 3 of end-stage renal disease. To date, none have died of a renal epithelial neoplasm. CONCLUSION The identification of unique renal neoplasia subtypes may provide important clues to establish a diagnosis of TSC, and in the somatic setting, this finding has important implications for accurate prognostication. These tumors tend to be indolent, and only 2 of 276 tumors in our study exhibited metastatic behavior. Our results support multidisciplinary management with a focus on preservation of renal function.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
| | - Rafael E Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | | | | | | | | | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|
77
|
Di Napoli C, Gennaro A, Lupica C, Falsaperla R, Leonardi R, Garozzo MT, Polizzi A, Praticò AD, Zanghì A, Ruggieri M. TSC1 and TSC2: Tuberous Sclerosis Complex and Its Related Epilepsy Phenotype. JOURNAL OF PEDIATRIC NEUROLOGY 2021. [DOI: 10.1055/s-0041-1727142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
AbstractTuberous sclerosis complex (TSC) is an autosomal dominant disorder characterized by a multisystemic involvement. In TSC, reduced function of TSC1 and TSC2 genes products (hamartin and tuberin, respectively) leads to an hyperactivation of the mechanistic target of rapamycin (mTOR) pathway and to a consequent cell growth dysregulation. In TSC patients, neurological and neuropsychiatric manifestations, especially epilepsy and neuropsychiatric comorbidities such as autism or intellectual disability, represent the most disabling features. In particular, epilepsy occurrs up to 80% of patients, is often drug resistant and is frequently associated with neurological impairment. Due to the burden of this morbidity, different treatment strategies have been proposed with the purpose to make patients epilepsy free, such as the use of different antiepileptic drugs like vigabatrin, carbamazepine, valproic acid, and levetiracetam. More recently, a mTOR inhibitor (i.e. everolimus) has showed promising results in terms of seizures reduction.
Collapse
Affiliation(s)
- Claudia Di Napoli
- Postgraduate Training Program in Genetics, Department of Biomedical and Biotechnological Sciences, Section of Genetics, University of Catania, Catania, Italy
| | - Alessia Gennaro
- Postgraduate Training Program in Genetics, Department of Biomedical and Biotechnological Sciences, Section of Genetics, University of Catania, Catania, Italy
| | - Carmelania Lupica
- Postgraduate Training Program in Genetics, Department of Biomedical and Biotechnological Sciences, Section of Genetics, University of Catania, Catania, Italy
| | - Raffaele Falsaperla
- Unit of Pediatrics and Pediatric Emergency, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
- Unit of Neonatal Intenstive Care and Neonatology, University Hospital “Policlinico Rodolico-San Marco,” Catania, Italy
| | - Roberta Leonardi
- Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, Unit of Rare Diseases of the Nervous System in Childhood, University of Catania, Catania, Italy
| | - Maria Teresa Garozzo
- Unit of Pediatrics and Pediatric Emergency, Hospital “Cannizzaro,” Catania, Italy
| | - Agata Polizzi
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
| | - Andrea D. Praticò
- Department of Clinical and Experimental Medicine, Section of Pediatrics and Child Neuropsychiatry, Unit of Rare Diseases of the Nervous System in Childhood, University of Catania, Catania, Italy
| | - Antonio Zanghì
- Department of General Surgery and Medical-Surgical Specialty, University of Catania, Catania, Italy
| | - Martino Ruggieri
- Chair of Pediatrics, Department of Educational Sciences, University of Catania, Catania, Italy
| |
Collapse
|
78
|
Yamamoto T, Kato T, Hatano K, Kawashima A, Ujike T, Fukuhara S, Kiuchi H, Imamura R, Ibuki N, Kiyotani K, Kurashige M, Morii E, Fujita K, Nonomura N, Uemura M. Genomic and Pathological Characterization of Multiple Renal Cell Carcinoma Regions in Patient With Tuberous Sclerosis Complex: A Case Report. Front Oncol 2021; 11:691996. [PMID: 34055654 PMCID: PMC8149899 DOI: 10.3389/fonc.2021.691996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Tuberous sclerosis complex is a genetic disorder characterized by facial angiofibromas, intellectual disability, epilepsy, and tumor formation in multiple organs, including the kidney. Renal cell carcinoma occurs in 2%–4% of patients with tuberous sclerosis complex, often developing multiply and bilaterally. Renal cell carcinoma associated with this genetic disorder may include complex tumor heterogeneity caused by the spatially different mutational landscape. Herein, we report the case of a female patient with tuberous sclerosis complex who developed multiple renal tumors. A 44-year-old female patient with tuberous sclerosis complex developed three different histological types of tumor—angiomyolipoma, clear cell renal cell carcinoma, and papillary renal cell carcinoma—in the left kidney at first renal cell carcinoma recurrence. The papillary renal cell carcinoma was morphologically atypical, indicating that its occurrence was associated with the genetic disorder. Furthermore, whole-exome sequencing revealed distinct patterns of somatic mutation in the three tumor types, and the atypical papillary renal cell carcinoma possessed a different mutational landscape than that of typical papillary renal cell carcinomas. Our findings indicate that tumors associated with tuberous sclerosis complex may be diagnosed with careful pathological examination. Furthermore, somatic mutation profiles of these tumors revealed their unique features, providing important information for further understanding the mechanism of multiple tumor development in patients with tuberous sclerosis complex.
Collapse
Affiliation(s)
- Tetsuya Yamamoto
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Hatano
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsunari Kawashima
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takeshi Ujike
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinichiro Fukuhara
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Kiuchi
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Naokazu Ibuki
- Department of Urology, Osaka Medical College, Osaka, Japan
| | - Kazuma Kiyotani
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masako Kurashige
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Motohide Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
79
|
Wu H, He D, Biswas S, Shafiquzzaman M, Zhou X, Charron J, Wang Y, Nayak BK, Habib SL, Liu H, Li B. mTOR Activation Initiates Renal Cell Carcinoma Development by Coordinating ERK and p38MAPK. Cancer Res 2021; 81:3174-3186. [PMID: 33863779 DOI: 10.1158/0008-5472.can-20-3979] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022]
Abstract
Renal cell carcinoma (RCC) mainly originates from renal proximal tubules. Intriguingly, disruption of genes frequently mutated in human RCC samples thus far has only generated RCC originated from other renal tubule parts in mouse models. This hampers our understanding of the pathogenesis of RCC. Here we show that mTOR signaling, often activated in RCC samples, initiates RCC development from renal proximal tubules. Ablation of Tsc1, encoding an mTOR suppressor, in proximal tubule cells led to multiple precancerous renal cysts. mTOR activation increased MEK1 expression and ERK activation, and Mek1 ablation or inhibition diminished cyst formation in Tsc1-deficient mice. mTOR activation also increased MKK6 expression and p38MAPK activation, and ablation of the p38α-encoding gene further enhanced cyst formation and led to RCC with clear cell RCC features. Mechanistically, Tsc1 deletion induced p53 and p16 expression in a p38MAPK-dependent manner, and deleting Tsc1 and Trp53 or Cdkn2a (encoding p16) enhanced renal cell carcinogenesis. Thus, mTOR activation in combination with inactivation of the p38MAPK-p53/p16 pathway drives RCC development from renal proximal tubules. Moreover, this study uncovers previously unidentified mechanisms by which mTOR controls cell proliferation and suggests the MEK-ERK axis to be a potential target for treatment of RCC. SIGNIFICANCE: Mouse modeling studies show that mTOR activation in combination with inactivation of the p38MAPK-p53/p16 axis initiates renal cell carcinoma that mimics human disease, identifying potential therapeutic targets for RCC treatment.
Collapse
Affiliation(s)
- Hongguang Wu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Dan He
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Soma Biswas
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Md Shafiquzzaman
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Zhou
- Institute of Traditional Chinese Medicine and Stem Cell Research, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jean Charron
- Centre de recherche du CHU de Québec-Université Laval (axe Oncologie), Centre de recherche sur le cancer de l'Université Laval, Université Laval, Québec, Canada
| | - Yibin Wang
- Department of Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Bijaya K Nayak
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Samy L Habib
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Huijuan Liu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China.
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China. .,Institute of Traditional Chinese Medicine and Stem Cell Research, School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,State Key Laboratory of Oncogenes and Related Genes, Bio-X-Renji Hospital Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
80
|
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
|
81
|
Garje R, Elhag D, Yasin HA, Acharya L, Vaena D, Dahmoush L. Comprehensive review of chromophobe renal cell carcinoma. Crit Rev Oncol Hematol 2021; 160:103287. [PMID: 33753250 DOI: 10.1016/j.critrevonc.2021.103287] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 11/27/2020] [Accepted: 02/27/2021] [Indexed: 12/12/2022] Open
Abstract
Chromophobe renal cell carcinoma (chRCC) is the third most common type of RCC with distinct biology compared to other kidney cancer subtypes. The heterogeneity between the RCC subtypes is associated with noticeable differences in tumor aggressiveness and risk for the development of metastatic disease. ChRCC is characterized by chromosomal aneuploidy, TP53, PTEN, and mitochondrial gene mutations. Though the therapeutic landscape of clear cell RCC (ccRCC) has significantly evolved over the past decade, limited progress has been seen in chRCC due to its infrequent incidence. In fact, the therapeutic approach for chRCC is often extrapolated from ccRCC treatments or studies that combine several forms of nccRCC subtypes. In the new era of genetic profiling of tumors and targeted therapeutics, this review describes the epidemiology, pathology, molecular characteristics, and current management with ongoing clinical trials for chRCC.
Collapse
Affiliation(s)
- Rohan Garje
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Department of Internal Medicine, University of Iowa, Iowa City, IA, United States.
| | - Dean Elhag
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Hesham A Yasin
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Luna Acharya
- Department of Internal Medicine, University of Iowa, Iowa City, IA, United States
| | - Daniel Vaena
- West Cancer Center and Research Institute, University of Tennessee, University of Iowa, United States
| | - Laila Dahmoush
- Department of Pathology and Urology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
82
|
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: 1.8] [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
|
83
|
Chen HIH, Lapadat R, Lastra RR, Biernacka A, Reeves W, Mueller J, Pambuccian SE, Barkan GA, Wojcik EM, Antic T. Accuracy of Subclassification and Grading of Renal Tumors on Fine Needle Aspiration Cytology Alone. Acta Cytol 2021; 65:140-149. [PMID: 33535202 DOI: 10.1159/000513065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 11/12/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Fine needle aspiration (FNA) of renal masses can distinguish between benign and malignant neoplasms in 73-94% of cases. Previous studies suggested the correct subclassification of renal cell carcinomas (RCCs) by cytomorphology can be achieved in up to 80% of cases. However, as RCCs become increasingly subclassified by molecular signatures, correct subclassification based on cytology alone is increasingly difficult. DESIGN Two FNA passes (2 stained with Diff-Quik® and 2 with the Papanicolaou method) were performed on all fresh nephrectomy specimens for a 1-year period. There were 30 cases in this study, with 29 primary renal tumors and 1 case of metastatic lung adenocarcinoma. Each case was assigned a random number and came with 2 slides (1 from each staining method). Eight cytopathologists were asked to provide a diagnosis and the World Health Organization/International Society of Urological Pathology (WHO/ISUP) grading if applicable. Fleiss' Kappa and Cohen's Kappa equations were used to look at inter-rater variability. RESULTS When compared to the surgical pathology diagnosis, the average percent correct diagnosis for all cytopathologist was 35%. Chromophobe RCCs had the best average percent accuracy at 72% followed by clearcell RCC at 48%. Average accuracy for grading RCCs was 40%. Inter-rater variability among the cytopathologists for all RCC diagnoses was fair with a Fleiss' Kappa coefficient of 0.28. For the WHO/ISUP grade, the weighted coefficient for each pathologist ranged from 0.11 to 0.45, ranging from fair to moderate, respectively. CONCLUSIONS Renal tumors are difficult to classify on cytopathology alone. Core needle biopsy and ancillary studies are necessary if diagnosis will change management.
Collapse
Affiliation(s)
- Heather I-Hsuan Chen
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA,
| | - Razvan Lapadat
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| | - Ricardo R Lastra
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| | - Anna Biernacka
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| | - Ward Reeves
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| | - Jeffrey Mueller
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| | - Stefan E Pambuccian
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois, USA
| | - Güliz A Barkan
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois, USA
| | - Eva M Wojcik
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois, USA
| | - Tatjana Antic
- Department of Pathology, The University of Chicago Hospitals, Chicago, Illinois, USA
| |
Collapse
|
84
|
Khan Y, Slattery TD, Pickering LM. Individualizing Systemic Therapies in First Line Treatment and beyond for Advanced Renal Cell Carcinoma. Cancers (Basel) 2020; 12:E3750. [PMID: 33322163 PMCID: PMC7764621 DOI: 10.3390/cancers12123750] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/28/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Therapeutic options for treating advanced renal cell cancer (RCC) are rapidly evolving. Vascular endothelial growth factor (VEGF)-directed therapy, predominantly VEGF receptor (VEGFr) tyrosine kinase inhibitors (TKIs) had been the most effective first line treatment since 2005 irrespective of International Metastatic RCC Database Consortium (IMDC) risk stratification. However, immune checkpoint inhibitors (ICI) have recently changed the treatment paradigm for advanced RCC particularly as the first-line systemic treatment modality. The combination of Ipilimumab and Nivolumab provides better disease control and long-term outcomes compared with the anti-VEGFr TKI Sunitinib for IMDC intermediate- to poor-risk patients and we now have the option of using ICI with TKI upfront for all IMDC risk groups. This poses a challenge for physicians, both to select the most suitable first line regimen and the most suitable subsequent therapy given the lack of data about sequencing in this setting. This treatment landscape is expected to become more complex with the emerging treatment options. Moreover, these therapeutic options cannot be generalized as significant variability exists between individual's disease biologies and their physiologies for handling treatment adverse effects. Notable efforts are being made to identify promising predictive biomarkers ranging from neo-antigen load to gene expression profiling. These biomarkers need prospective validation to justify their utility in clinical practice and in treatment decision making. This review article discusses various clinicopathological characteristics that should be carefully evaluated to help select appropriate treatment and discusses the current status of biomarker-based selection.
Collapse
Affiliation(s)
| | | | - Lisa M. Pickering
- The Royal Marsden Hospital NHS Foundation Trust, Fulham Road, London SW3 6JJ, UK; (Y.K.); (T.D.S.)
| |
Collapse
|
85
|
Wu Y, Li H, Yu X, Lin M, Chen Y. A Case of Tuberous Sclerosis Complex with Lymphangioleiomyomatosis and Renal Cell Carcinoma. Onco Targets Ther 2020; 13:12421-12426. [PMID: 33293833 PMCID: PMC7719313 DOI: 10.2147/ott.s276627] [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: 08/13/2020] [Accepted: 11/15/2020] [Indexed: 12/02/2022] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a rare disease affecting young women, which occurs sporadically or in patients with tuberous sclerosis complex (TSC). The main manifestations of TSC in the kidney include cysts and angiomyolipoma (AML). Although renal cell carcinoma (RCC) is not a manifestation of TSC, it has a 2–4% incidence in TSC patients. Furthermore, LAM is rare in patients with RCC. Herein, we present a case of a 40-year-old woman with LAM and RCC in the right kidney. We checked for mutations in the TSC1 and TSC2 genes from both blood and kidney lesions and found a heterozygous mutation of c.1717–30G> A in intron 16 of TSC2 gene. In TSC patients, the diagnosis of RCC is challenging because the cancer is rare, and it is often difficult to distinguish it from AML with conventional imaging techniques. Therefore, it is recommended that patients with TSC undergo renal imaging follow-ups annually for kidney masses.
Collapse
Affiliation(s)
- Yun Wu
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Hongru Li
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Xiaoli Yu
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Ming Lin
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| | - Yusheng Chen
- Department of Respiratory Medicine, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, People's Republic of China
| |
Collapse
|
86
|
Molecular characterization of chromophobe renal cell carcinoma reveals mTOR pathway alterations in patients with poor outcome. Mod Pathol 2020; 33:2580-2590. [PMID: 32616874 DOI: 10.1038/s41379-020-0607-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 06/12/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
Chromophobe renal cell carcinoma (chRCC) is a histologically and molecularly distinct class of rare renal tumor. TCGA studies revealed low mutational burden, with only TP53 and PTEN recurrently mutated, and discovered alterations in TERT promoter and in the electron transport chain Complex I genes. However, knowledge on drug targetable genes is limited and treatments at metastatic stage do not follow a molecular rationale. In a large series of 92 chRCC enriched with metastatic cases, we performed an in-depth characterization of mTOR pathway alterations through targeted NGS and immunohistochemistry (IHC) of phospho-S6, tuberin, and PTEN. Mutations in mitochondria, telomere maintenance and other renal cancer related genes and p53 IHC, were also assessed. The impact on metastasis development and disease specific survival was determined, using TCGA-KICH series (n = 65) for validation. mTOR pathway mutations (MTOR, TSC1, TSC2) were present in 17% of primary tumors, most of them being classified as pathogenic. Mutations were associated with positive IHC staining of phospho-S6 and PTEN (P = 0.009 and P = 0.001, respectively) and with chRCC eosinophilic variant (P = 0.039), supporting a biological relevance of the pathway. mTOR pathway mutations were associated with worse clinical outcomes. Survival analysis gave a hazard ratio of 5.5 (P = 0.027), and this association was confirmed in TCGA-KICH (HR = 10.3, P = 0.006). TP53 mutations were enriched in metastatic cases (P = 0.018), and mutations in telomere maintenance genes showed a trend in the same direction. p53 IHC staining pattern was associated with the underlying TP53 defect, and negative PTEN IHC staining (82% of cases) suggested PTEN loss as a chRCC hallmark. In conclusion, our study provides with novel genomic knowledge in chRCC and identifies novel markers of poor survival. Furthermore, this is the first study showing that mTOR pathway mutations correlate with poor prognosis, and may help to identify patients with increased sensitivity to mTOR inhibitors.
Collapse
|
87
|
Angiomyolipoma of the kidney: from simple hamartoma to complex tumour. Pathology 2020; 53:129-140. [PMID: 33131798 DOI: 10.1016/j.pathol.2020.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022]
Abstract
Angiomyolipoma is the most common mesenchymal tumour of the kidney, even if for a long time it has been viewed as a hamartoma rather than a neoplasm. It belongs to a family of neoplasms, named PEComa, characterised by the constant presence of perivascular epithelioid cells that co-express smooth muscle and melanogenesis markers. Angiomyolipoma can occur in patients with tuberous sclerosis, a hereditary syndrome due to the alteration of TSC1 or TSC2 genes, or sporadically. Angiomyolipoma and its variants are indolent tumours; however, some epithelioid angiomyolipomas/pure epithelioid PEComas are aggressive, and criteria for malignancy have been proposed to identify those cases. Although typical angiomyolipoma is a straightforward diagnosis, pathologists should be aware of the wide morphological spectrum of its variants which could be tricky in routine clinical practice and could require immunohistochemical analysis for resolution. The differential diagnosis may range from an inflammatory process (for instance xanthogranulomatous pyelonephritis) to the most common renal cancers and sarcomas. The immunoexpression of melanogenesis markers (HMB45 and Melan-A) and cathepsin K is extremely helpful in the majority of cases. Recently, a subset of epithelioid angiomyolipoma/pure epithelioid PEComa harbouring TFE3 gene fusions has been described, raising questions about its relationship with the family of perivascular epithelioid cell tumour. The activation of the mTOR pathway due to genetic alterations of tuberous sclerosis complex in TSC1 or TSC2 genes in angiomyolipoma has also been reported as well as the subsequent therapeutic implications.
Collapse
|
88
|
Hamroun A, Lenain R, Provôt F, Maanaoui M, Lionet A. An unusual case of multiple bilateral kidney tumors. J Nephrol 2020; 34:1783-1784. [PMID: 33113122 DOI: 10.1007/s40620-020-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Aghilès Hamroun
- Nephrology, Dialysis, and Kidney Transplantation Department, Lille University, Regional and University Hospital Center of Lille, 59000, Lille, France. .,Center for Research in Epidemiology and Population Health (CESP), Clinical Epidemiology Team, National Institute of Health and Medical Research, Villejuif, France.
| | - Rémi Lenain
- Nephrology, Dialysis, and Kidney Transplantation Department, Lille University, Regional and University Hospital Center of Lille, 59000, Lille, France
| | - François Provôt
- Nephrology, Dialysis, and Kidney Transplantation Department, Lille University, Regional and University Hospital Center of Lille, 59000, Lille, France
| | - Mehdi Maanaoui
- Nephrology, Dialysis, and Kidney Transplantation Department, Lille University, Regional and University Hospital Center of Lille, 59000, Lille, France.,INSERM U1190, Translational Research for Diabetes, Lille, France
| | - Arnaud Lionet
- Nephrology, Dialysis, and Kidney Transplantation Department, Lille University, Regional and University Hospital Center of Lille, 59000, Lille, France
| |
Collapse
|
89
|
Eosinophilic Renal Cell Tumors With a TSC and MTOR Gene Mutations Are Morphologically and Immunohistochemically Heterogenous: Clinicopathologic and Molecular Study. Am J Surg Pathol 2020; 44:943-954. [PMID: 32091432 DOI: 10.1097/pas.0000000000001457] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Eosinophilic renal neoplasms have a wide spectrum of histologic presentations, and several studies have demonstrated a subtype of renal cell carcinomas (RCCs) associated with the tuberous sclerosis complex (TSC)/mammalian target of rapamycin pathway. A review of our institutional archives led to the identification of 18 cases of renal eosinophilic tumors with unusual morphology. Immunohistochemical analysis demonstrated that these could be separated into 3 groups: group 1 had solid architecture and morphology similar to chromophobe RCC but was negative for CK20 and vimentin, and had weak focal staining for CK7 and P504S; group 2 had solid architecture and morphology similar to either renal oncocytoma or chromophobe RCC, eosinophilic variant and had diffuse staining of CK7 and P504S, absent to weak staining of CK20, and negative staining for vimentin; and group 3 had solid, cystic and papillary architecture and was negative for CK7, except for 1 case, along with moderate to strong staining of CK20, P504S, and vimentin. The cases were then sent for next-generation sequencing to determine whether molecular pathogenic variants were present. In group 1, all 3 cases had mutations in TSC2. In group 2, pathogenic variants were identified in 3 genes: TSC1, TSC2, and MTOR. In group 3, genetic alterations and pathogenic variants were identified in TSC1 and TSC2. Our results support TSC/MTOR-associated neoplasms as a distinct group that exhibits heterogenous morphology and immunohistochemical staining.
Collapse
|
90
|
Griffin BB, Lin X. Cytomorphologic analysis of clear cell papillary renal cell carcinoma: Distinguishing diagnostic features. Cancer Cytopathol 2020; 129:192-203. [PMID: 33036062 DOI: 10.1002/cncy.22355] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Clear cell papillary renal cell carcinoma (CCPRCC) shares histomorphologic and immunophenotypic features with clear cell RCC (CCRCC) and papillary RCC (PRCC). METHODS We compared the cytomorphology, immunoprofile, and clinical management of CCPRCC (n = 18), CCRCC (n = 20), and PRCC (n = 18). RESULTS Useful cytomorphologic features for comparing CCPRCC with CCRCC include 3-dimensional clusters (72% vs 0%), papillae (50% vs 0%) and sheets (22% vs 70%), vasculature (papillary vs traversing), naked nuclei (17% vs 100%), prominent nucleoli (0% vs 65%), and amount of cytoplasm (small vs large). Useful cytomorphologic features for comparing CCPRCC with PRCC include sheets (22% vs 61%), naked nuclei (17% vs 67%), nuclear grooves (5% vs 67%) and inclusions (17% vs 67%), and pigmented cytoplasm (17% vs 83%). At on-site evaluation, 16 of 18 (86%) CCPRCC specimens were deemed adequate, with suspicion for CCPRCC in 5 of 16 (31%) cases. Core histology of CCPRCC showed low-grade malignant cells in nests (67%), tubules (100%), and papillae (72%), frequently in myxohyaline stroma (67%). Immunostains demonstrated expression of cytokeratin 7 (CK7; 100%), carbonic anhydrase IX (CA IX; 100%, cup-like), CD10 (53%, reverse cup-like), and α-methylacyl-CoA racemase (AMACR; 35%). Among 18 CCPRCC patients, 9 (50%) underwent nephrectomy, 5 (28%) underwent cryo-ablation, and 4 (22%) were under surveillance with serial imaging. CONCLUSION Certain morphologic features represent diagnostic criteria of CCPRCC in cytology specimens and help distinguish CCPRCC from CCRCC and PRCC. Immunostaining patterns with CK7, CA IX, CD10, and AMACR can confirm the diagnosis. Delineating CCPRCC from more biologically aggressive RCC types in cytology specimens enhances presurgical and clinical management of patients given CCPRCC's low-grade, indolent behavior.
Collapse
Affiliation(s)
- Brannan B Griffin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiaoqi Lin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
91
|
Smith PS, West H, Whitworth J, Castle B, Sansbury FH, Warren AY, Woodward ER, Tischkowitz M, Maher ER. Pathogenic germline variants in patients with features of hereditary renal cell carcinoma: Evidence for further locus heterogeneity. Genes Chromosomes Cancer 2020; 60:5-16. [PMID: 32830346 DOI: 10.1002/gcc.22893] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/12/2020] [Accepted: 08/19/2020] [Indexed: 11/08/2022] Open
Abstract
Inherited renal cell carcinoma (RCC) is associated with multiple familial cancer syndromes but most individuals with features of non-syndromic inherited RCC do not harbor variants in the most commonly tested renal cancer predisposition genes (CPGs). We investigated whether undiagnosed cases might harbor mutations in CPGs that are not routinely tested for by testing 118 individuals with features suggestive of inherited RCC (family history of RCC, two or more primary RCC aged <60 years, or early onset RCC ≤46 years) for the presence of pathogenic variants in a large panel of CPGs. All individuals had been prescreened for pathogenic variants in the major RCC genes. We detected pathogenic or likely pathogenic (P/LP) variants of potential clinical relevance in 16.1% (19/118) of individuals, including P/LP variants in BRIP1 (n = 4), CHEK2 (n = 3), MITF (n = 1), and BRCA1 (n = 1). Though the power to detect rare variants was limited by sample size the frequency of truncating variants in BRIP1, 4/118, was significantly higher than in controls (P = 5.92E-03). These findings suggest that the application of genetic testing for larger inherited cancer gene panels in patients with indicators of a potential inherited RCC can increase the diagnostic yield for P/LP variants. However, the clinical utility of such a diagnostic strategy requires validation and further evaluation and in particular, confirmation of rarer RCC genotype-phenotype associations is required.
Collapse
Affiliation(s)
- Philip S Smith
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Hannah West
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - James Whitworth
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Bruce Castle
- Peninsula Clinical Genetics Service, Royal Devon and Exeter NHS Foundation Trust, Royal Devon and Exeter Hospital (Heavitree), Exeter, UK University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Francis H Sansbury
- Peninsula Clinical Genetics Service, Royal Devon and Exeter NHS Foundation Trust, Royal Devon and Exeter Hospital (Heavitree), Exeter, UK University of Exeter Medical School, University of Exeter, Exeter, UK.,All Wales Medical Genomics Service, Cardiff and Vale University Health Board, Institute of Medical Genetics, University Hospital of Wales, Cardiff, UK
| | - Anne Y Warren
- Department of Histopathology, Cambridge University NHS Foundation Trust and Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Emma R Woodward
- Manchester Centre for Genomic Medicine and NW Laboratory Genetics Hub, Manchester University Hospitals NHS Foundation Trust, and Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Health Innovation Manchester, Manchester, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Eamonn R Maher
- Department of Medical Genetics, University of Cambridge and NIHR Cambridge Biomedical Research Centre, and Cancer Research UK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
92
|
Kingswood JC, Belousova E, Benedik MP, Carter T, Cottin V, Curatolo P, Dahlin M, D'Amato L, Beaure d'Augères G, de Vries PJ, Ferreira JC, Feucht M, Fladrowski C, Hertzberg C, Jozwiak S, Lawson JA, Macaya A, Marques R, Nabbout R, O'Callaghan F, Qin J, Sander V, Shah S, Takahashi Y, Touraine R, Youroukos S, Zonnenberg B, Jansen AC, Sauter M. Renal Manifestations of Tuberous Sclerosis Complex: Key Findings From the Final Analysis of the TOSCA Study Focussing Mainly on Renal Angiomyolipomas. Front Neurol 2020; 11:972. [PMID: 33041968 PMCID: PMC7526256 DOI: 10.3389/fneur.2020.00972] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/24/2020] [Indexed: 12/18/2022] Open
Abstract
Renal angiomyolipomas are one of the most common renal manifestations in patients with tuberous sclerosis complex (TSC), with potentially life-threatening complications and a poor prognosis. Despite the considerable progress in understanding TSC-associated renal angiomyolipomas, there are no large scale real-world data. The aim of our present study was to describe in detail the prevalence and outcome of renal angiomyolipomas in patients with TSC, enrolled into the TuberOus SClerosis registry to increase disease Awareness (TOSCA) from 170 sites across 31 countries worldwide. We also sought to evaluate the relationship of TSC-associated renal angiomyolipomas with age, gender and genotype. The potential risk factors for renal angiomyolipoma-related bleeding and chronic kidney disease (CKD) were studied in patients who participated in the TOSCA renal angiomyolipoma substudy. Of the 2,211 eligible patients, 1,062 (48%) reported a history of renal angiomyolipomas. The median age of TSC diagnosis for the all subjects (n = 2,211) was 1 year. The median age of diagnosis of renal angiomyolipoma in the 1,062 patients was 13 years. Renal angiomyolipomas were significantly more prevalent in female patients (p < 0.0001). Rates of angiomyolipomas >3 cm (p = 0.0119), growing lesions (p = 0.0439), and interventions for angiomyolipomas (p = 0.0058) were also higher in females than males. Pre-emptive intervention for renal angiomyolipomas with embolisation, surgery, or mammalian target of rapamycin (mTOR) inhibitor may have abolished the gender difference in impaired renal function, hypertension, and other complications. The rate of interventions for angiomyolipomas was less common in children than in adults, but interventions were reported in all age groups. In the substudy of 76 patients the complication rate was too low to be useful in predicting risk for more severe CKD. In addition, in this substudy no patient had a renal hemorrhage after commencing on an mTOR inhibitor. Our findings confirmed that renal angiomyolipomas in subjects with TSC1 mutations develop on average at the later age, are relatively smaller in size and less likely to be growing; however, by age 40 years, no difference was observed in the percentage of patients with TSC1 and TSC2 mutations needing intervention. The peak of appearance of new renal angiomyolipomas was observed in patients aged between 18 and 40 years, but, given that angiomyolipomas can occur later, lifelong surveillance is necessary. We found that pre-emptive intervention was dramatically successful in altering the outcome compared to historical controls; with high pre-emptive intervention rates but low rates of bleeding and other complications. This validates the policy of surveillance and pre-emptive intervention recommended by clinical guidelines.
Collapse
Affiliation(s)
- J Chris Kingswood
- Cardiology Clinical Academic Group, Molecular and Clinical Sciences Research Centre, St Georges University of London, London, United Kingdom
| | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Mirjana P Benedik
- Child Neurology Department, SPS Pediatrična Klinika, Ljubljana, Slovenia
| | - Tom Carter
- TSA Tuberous Sclerosis Association, Nottingham, United Kingdom
| | - Vincent Cottin
- Hôpital Louis Pradel, Claude Bernard University Lyon 1, Lyon, France
| | - Paolo Curatolo
- Child Neurology and Psychiatry Unit, Systems Medicine Department, Tor Vergata University Hospital, Rome, Italy
| | - Maria Dahlin
- Neuropediatric Department, Astrid Lindgren Children's Hospital, Stockholm, Sweden
| | | | | | - Petrus J de Vries
- Division of Child and Adolescent Psychiatry, University of Cape Town, Cape Town, South Africa
| | - José C Ferreira
- Neurology Department, Centro Hospitalar Lisboa Ocidental, Lisbon, Portugal
| | - Martha Feucht
- Medical University of Vienna, Universitätsklinik für Kinder-und Jugendheilkunde, Affiliated Partner of the ERN EpiCARE, Vienna, Austria
| | - Carla Fladrowski
- Associazione Sclerosi Tuberosa ONLUS, Milan, Italy.,European Tuberous Sclerosis Complex Association, Dattein, Germany
| | - Christoph Hertzberg
- Zentrum für Neuropädiatrie und Sozialpädiatrie, Vivantes-Klinikum Neukölln, Berlin, Germany
| | - Sergiusz Jozwiak
- Department of Child Neurology, Medical University of Warsaw, Warsaw, Poland.,Department of Neurology and Epileptology, The Children's Memorial Health Institute, Warsaw, Poland
| | - John A Lawson
- The Tuberous Sclerosis Multidisciplinary Management Clinic, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Alfons Macaya
- Pediatric Neurology Section, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Ruben Marques
- Novartis Farma S.p.A., Origgio, Italy.,Institute of Biomedicine (IBIOMED), University of Leon, León, Spain
| | - Rima Nabbout
- Department of Pediatric Neurology, Necker Enfants Malades Hospital, Paris Descartes University, Paris, France
| | - Finbar O'Callaghan
- Institute of Child Health, University College London, London, United Kingdom
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital (PKUPH), Beijing, China
| | - Valentin Sander
- Department of Neurology and Rehabilitation, Tallinn Children Hospital, Tallinn, Estonia
| | - Seema Shah
- Novartis Healthcare Pvt. Ltd., Hyderabad, India
| | - Yukitoshi Takahashi
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorders, Shizuoka, Japan
| | - Renaud Touraine
- Department of Genetics, CHU-Hôpital Nord, Saint-Étienne, France
| | - Sotiris Youroukos
- First Department of Paediatrics, Athens University, St. Sophia Children's Hospital, Athens, Greece
| | - Bernard Zonnenberg
- Department of Internal Medicine, University Medical Center, Utrecht, Netherlands
| | - Anna C Jansen
- Pediatric Neurology Unit, Department of Pediatrics, UZ Brussel VUB, Brussels, Belgium
| | | |
Collapse
|
93
|
Abstract
Tuberous sclerosis complex (TSC) is a genetic condition caused by a mutation in either the TSC1 or TSC2 gene. Disruption of either of these genes leads to impaired production of hamartin or tuberin proteins, leading to the manifestation of skin lesions, tumors, and seizures. TSC can manifest in multiple organ systems with the cutaneous and renal systems being the most commonly affected. These manifestations can secondarily lead to the development of hypertension, chronic kidney disease, and neurocognitive declines. The renal pathologies most commonly seen in TSC are angiomyolipoma, renal cysts, and less commonly, oncocytomas. In this review, we highlight the current understanding on the renal manifestations of TSC along with current diagnosis and treatment guidelines.
Collapse
|
94
|
Gournay M, Dugay F, Belaud-Rotureau MA, Peyronnet B, Mathieu R, Verhoest G, Bensalah K, Odent S, Denizeau P, Vigneau C, Morini A, Rioux-Leclercq N, Kammerer-Jacquet SF. Renal cell carcinoma with leiomyomatous stroma in tuberous sclerosis complex: a distinct entity. Virchows Arch 2020; 478:793-799. [PMID: 32845354 DOI: 10.1007/s00428-020-02910-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 01/19/2023]
Abstract
Renal cell carcinoma with leiomyomatous stroma (RCCLS) is an emerging entity frequently associated with tuberous sclerosis complex (TSC). We described herein a series of RCCLS in TSC patients at pathological and cytogenetic levels. Three male patients with TSC and RCCLS were identified between 2000 and 2019 at the University Hospital of Rennes. Histologically, the architecture was tubulo-papillary with thick bundles of smooth muscle cells. The tumor cells showed clear cytoplasm with eosinophilic globules. The immunohistochemical profile was identical with an intense positivity of CK7, CAIX, and CD10 and a heterogeneous positivity of CK20. SDHB was low but positive and TFE3 was not expressed. Comparative genomic hybridization (CGH) did not show any quantitative chromosome abnormality. No recurrence was observed with a median follow-up of 4 years. RCCLS in TSC patients has morphological, immunohistochemical, and cytogenetic distinct features that could constitute a distinct entity and a sentinel manifestation for the diagnosis of TSC.
Collapse
Affiliation(s)
- Marjorie Gournay
- Department of Pathology, University Hospital, 2 rue Henri le Guilloux, 35000, Rennes, France.
| | - Frédéric Dugay
- Department of Cytogenetics, University Hospital, 35000, Rennes, France
| | | | - Benoit Peyronnet
- Department of Urology, University Hospital, 35000, Rennes, France
| | - Romain Mathieu
- Department of Urology, University Hospital, 35000, Rennes, France
| | - Gregory Verhoest
- Department of Urology, University Hospital, 35000, Rennes, France
| | - Karim Bensalah
- Department of Urology, University Hospital, 35000, Rennes, France
| | - Sylvie Odent
- Department of Genetic, University Hospital, 35000, Rennes, France
| | | | - Cécile Vigneau
- Department of Nephrology, University Hospital, 35000, Rennes, France
| | - Aurélien Morini
- Department of Pathology, Georges Pompidou European Hospital, Paris, France
| | - Nathalie Rioux-Leclercq
- Department of Pathology, University Hospital, 2 rue Henri le Guilloux, 35000, Rennes, France
| | | |
Collapse
|
95
|
"Renal Cell Carcinoma With Leiomyomatous Stroma" Harbor Somatic Mutations of TSC1, TSC2, MTOR, and/or ELOC (TCEB1): Clinicopathologic and Molecular Characterization of 18 Sporadic Tumors Supports a Distinct Entity. Am J Surg Pathol 2020; 44:571-581. [PMID: 31850909 DOI: 10.1097/pas.0000000000001422] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma with (angio) leiomyomatous stroma (RCCLMS) is included as a provisional entity in the 2016 World Health Organization (WHO) classification of renal epithelial neoplasia; however, debate remains whether it represents a distinct entity or a heterogenous group of renal cell carcinomas (RCCs) with overlapping morphology. Also, its relationship to similar tumors occurring in the setting of tuberous sclerosis complex (TSC) is not fully addressed. We analyzed the clinicopathologic, immunohistochemical, and molecular characteristics of 23 sporadic RCCs associated with smooth muscle stroma and classified them into 2 groups, independent of molecular results: (1) RCCLMS (n=18) and (2) clear cell renal cell carcinoma (CCRCC) (n=5). The classification of a case as "RCCLMS" was based on morphologic comparison with 5 "index" RCCs from 3 patients with TSC showing similar features and the presence of diffuse CK7 expression. To investigate mutational and copy number alterations, a 170-gene solid tumor panel was utilized to sequence 14 RCCLMSs and control of 5 CCRCCs. Also, 4 RCCLMSs, suspicious for chromosome 8 monosomy, were further evaluated by a broader 479 gene sequencing panel that included ELOC (also referred to as TCEB1). Clinical information and follow-up data were obtained from electronic medical records. The mean age of patients with RCCLMS was 52 years (range, 33 to 69) with male:female ratio of 1:2. Macroscopically, all tumors were solitary and predominantly (82%) tan/red, circumscribed, and solid. The average tumor size was 2.3 cm (range, 1.1 to 4.5). Microscopically, the distinctive feature included tumor nodules of elongated and frequently branching tubules lined by cells with voluminous clear to mildly eosinophilic cytoplasm (100%), separated by focal to prominent smooth muscle stroma. Additional frequently identified features included: biphasic pattern of collapsed acini surrounding tubules with voluminous cytoplasm (50%), focal papillary architecture (39%), peritumoral lymphoid aggregates (39%), and hemosiderin-laden macrophages (33%). All 11 (100%) RCCLMSs with available staging information were pT1; 78% were WHO/International Society of Urologic Pathology (ISUP) grade 2 and 22% grade 3. Immunophenotypically, RCCLMSs were characterized by diffuse CK7, CAM5.2 and CD10 reactivity (100%). All patients with available follow-up (n=10) were alive and without disease progression after a mean and median follow-up of 25.2 (range: 1 to 58) and 25 months, respectively. The molecular results showed recurrent mutations in all RCCLMS: TSC1 (4), TSC2 (4), MTOR (6), and/or ELOC (2). Five control CCRCCs demonstrated primary alterations in VHL gene, while all 14 RCCLMS cases tested had intact VHL gene. Of 2 RCCLMSs with confirmed monosomy 8, 1 showed a hotspot ELOC mutation without TSC/MTOR mutations, and 1 showed a previously undescribed 3-bp in-frame ELOC deletion, along with a truncating TSC1 mutation. In conclusion, RCCLMS, as defined herein, harbors recurrent mutations of TSC1/TSC2, MTOR, and/or ELOC, consistent with hyperactive MTOR complex. Our findings argue that these tumors represent the sporadic counterpart to morphologically identical tumors occurring in TSC patients. Finally, the data support that RCCLMS is a novel subtype of RCC with unique morphologic, immunohistochemical, and molecular characteristics that is distinct from CCRCC and clear cell-papillary RCC.
Collapse
|
96
|
Williamson SR, Gill AJ, Argani P, Chen YB, Egevad L, Kristiansen G, Grignon DJ, Hes O. Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers: III: Molecular Pathology of Kidney Cancer. Am J Surg Pathol 2020; 44:e47-e65. [PMID: 32251007 PMCID: PMC7289677 DOI: 10.1097/pas.0000000000001476] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Renal cell carcinoma (RCC) subtypes are increasingly being discerned via their molecular underpinnings. Frequently this can be correlated to histologic and immunohistochemical surrogates, such that only simple targeted molecular assays, or none at all, are needed for diagnostic confirmation. In clear cell RCC, VHL mutation and 3p loss are well known; however, other genes with emerging important roles include SETD2, BAP1, and PBRM1, among others. Papillary RCC type 2 is now known to include likely several different molecular entities, such as fumarate hydratase (FH) deficient RCC. In MIT family translocation RCC, an increasing number of gene fusions are now described. Some TFE3 fusion partners, such as NONO, GRIPAP1, RBMX, and RBM10 may show a deceptive fluorescence in situ hybridization result due to the proximity of the genes on the same chromosome. FH and succinate dehydrogenase deficient RCC have implications for patient counseling due to heritable syndromes and the aggressiveness of FH-deficient RCC. Immunohistochemistry is increasingly available and helpful for recognizing both. Emerging tumor types with strong evidence for distinct diagnostic entities include eosinophilic solid and cystic RCC and TFEB/VEGFA/6p21 amplified RCC. Other emerging entities that are less clearly understood include TCEB1 mutated RCC, RCC with ALK rearrangement, renal neoplasms with mutations of TSC2 or MTOR, and RCC with fibromuscular stroma. In metastatic RCC, the role of molecular studies is not entirely defined at present, although there may be an increasing role for genomic analysis related to specific therapy pathways, such as for tyrosine kinase or MTOR inhibitors.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mutation
- Neoplasm Metastasis
- Neoplastic Syndromes, Hereditary/diagnosis
- Neoplastic Syndromes, Hereditary/genetics
- Neoplastic Syndromes, Hereditary/metabolism
- Neoplastic Syndromes, Hereditary/pathology
- Pathology, Clinical
- Pathology, Molecular
- Prognosis
- Societies, Medical
- Urology
Collapse
Affiliation(s)
- Sean R Williamson
- Department of Pathology and Laboratory Medicine and Henry Ford Cancer Institute, Henry Ford Health System
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - Anthony J Gill
- NSW Health Pathology, Department of Anatomical Pathology
- Cancer Diagnosis and Pathology Research Group, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Pedram Argani
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ying-Bei Chen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - David J Grignon
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN
| | - Ondrej Hes
- Department of Pathology, Charles University, Medical Faculty and Charles University Hospital Plzen, Pilsen, Czechia
| |
Collapse
|
97
|
Williamson SR, Cardili L, Whiteley LJ, Sanchez J, Kis O. Sclerosing TSC1 mutated renal cell carcinoma: An unusual pattern mimicking MITF family translocation renal cell carcinoma. Genes Chromosomes Cancer 2020; 59:591-594. [PMID: 32418252 DOI: 10.1002/gcc.22860] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/07/2020] [Accepted: 05/13/2020] [Indexed: 01/12/2023] Open
Abstract
The tuberous sclerosis genes and MTOR are increasingly being found to have important roles in novel subtypes of renal cancer, particularly emerging entities eosinophilic solid and cystic renal cell carcinoma (RCC) and high-grade oncocytic renal tumor (HOT)/RCC with eosinophilic and vacuolated cytoplasm. We report a unique renal neoplasm in a 66-year-old woman that initially mimicked MITF family translocation RCC due to mixed clear and eosinophilic cells, extensive stromal hyalinization, and psammoma bodies, yet which was negative for TFE3 and TFEB fluorescence in situ hybridization and a next generation sequencing (NGS) gene fusion assay. Cytoplasmic stippling triggered consideration of TSC-associated neoplasms, and a targeted NGS assay revealed a variant in exon 21 of TSC1 resulting in c.2626G>T p.(Glu876*) truncating mutation. This report adds to the morphologic spectrum of TSC-related renal neoplasms, including prominent stromal hyalinization as a potentially deceptive pattern. Due to the overlap in cytoplasmic stippling between eosinophilic solid and cystic RCC and HOT/RCC with eosinophilic and vacuolated cytoplasm, it is debatable which category this example would best fit. Further understanding of these entities and other renal neoplasms with alterations in the TSC genes will elucidate whether they should be considered a family of tumors.
Collapse
Affiliation(s)
- Sean R Williamson
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, Michigan, USA.,Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Leonardo Cardili
- Department of Pathology, Cancer Institute of the State of São Paulo, São Paulo, Brazil
| | - Lisa J Whiteley
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, Michigan, USA
| | - Jessica Sanchez
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, Michigan, USA
| | - Olena Kis
- Department of Pathology and Laboratory Medicine, Henry Ford Health System, Detroit, Michigan, USA
| |
Collapse
|
98
|
Walker SM, Gautam R, Turkbey B, Malayeri A, Choyke PL. Update on Hereditary Renal Cancer and Imaging Implications. Radiol Clin North Am 2020; 58:951-963. [PMID: 32792126 DOI: 10.1016/j.rcl.2020.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Up to 8% of renal cancers are thought to have a hereditary component. Several hereditary renal cancer syndromes have been identified over the last few decades. It is important for the radiologist to be aware of findings associated with hereditary renal cancer syndromes to detect tumors early, enroll patients in appropriate surveillance programs, and improve outcomes for the patient and affected family members. This review discusses from a radiologist's perspective well-known hereditary renal cancer syndromes and emerging genetic mutations associated with renal cancer that are less well characterized, focusing on imaging features and known associations.
Collapse
Affiliation(s)
- Stephanie M Walker
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Rabindra Gautam
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Ashkan Malayeri
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| |
Collapse
|
99
|
Zhang L, Henske EP. Chromophobe renal cell carcinoma: New genetic and metabolic insights. Urol Oncol 2020; 38:678-681. [PMID: 32444178 DOI: 10.1016/j.urolonc.2020.04.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/08/2020] [Accepted: 04/30/2020] [Indexed: 11/18/2022]
Abstract
Chromophobe renal cell carcinoma (chRCC) represents 5% of all RCC. ChRCC appears to arise from the distal nephron, in contrast to clear cell RCC that arises from the proximal nephron. ChRCC is distinctive because the tumor cells contain abundant abnormal mitochondria, and frequently have loss of one copy of multiple chromosomes, with a low rate of somatic mutations. Here, we focus on recent discoveries related to genetic and metabolic factors that may promote the progression of chRCC and summarize how these findings may relate to the prognosis and treatment of chRCC.
Collapse
Affiliation(s)
- Long Zhang
- Brigham and Women's Hospital, Boston, MA
| | | |
Collapse
|
100
|
Verkarre V, Morini A, Denize T, Ferlicot S, Richard S. [Hereditary kidney cancers: The pathologist's view in 2020]. Ann Pathol 2020; 40:148-167. [PMID: 32197858 DOI: 10.1016/j.annpat.2020.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/23/2022]
Abstract
Hereditary predispositions to adult kidney tumors involve around 5% of tumors and include a dozen of autosomal dominant syndromes. The most frequent tumors encountered in these setting are clear cell renal cell carcinomas, papillary renal cell carcinomas, chromophobe renal cell carcinomas and angiomyolipomas. Their detection is essential in order to adapt individual care and perform genetic screening of at-risk relatives, especially in the national french network PREDIR, labeled by the National Cancer Institute and dedicated to hereditary predispositions to kidney tumors. Targeted genetic analysis, which was guided in particular by the renal tumor subtype, has recently evolved into genetic analysis using panels of genes. Pathologist contribution's remains however central in the diagnosis of hereditary forms since we currently have immunohistochemical biomarkers that allow us to diagnose two specifically hereditary entities: hereditary leiomyomatosis and renal cell carcinoma associated-renal cell carcinoma, associated with a loss of fumarate hydratase and succinate dehydrogenase-deficient renal cell carcinoma associated with a loss of succinate deshydrogenase B expression. These diagnoses must however be confirmed by the identification of pathogenic germline variation in the corresponding genes. Improvement of kidney tumors characterization has also lead to identify new subtypes, expanding the algorithm of renal tumors associated with hereditary setting. Here we aim to review all subtypes of adult renal tumors encountered in predisposition syndromes.
Collapse
Affiliation(s)
- Virginie Verkarre
- Service d'anatomie pathologique, université de Paris, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris-Centre, 20, rue Leblanc, 75015 Paris, France; Inserm U970, équipe labellisée par la Ligue contre le cancer, PARCC, université de Paris, Paris, France; Réseau national de référence pour cancers rares de l'adulte PREDIR (« Maladie de von Hippel-Lindau et prédispositions héréditaires au cancer rénal ») labellisée par l'Institut national du cancer, université Paris Saclay, Assistance publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France.
| | - Aurélien Morini
- Service d'anatomie pathologique, université de Paris, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris-Centre, 20, rue Leblanc, 75015 Paris, France
| | - Thomas Denize
- Service d'anatomie pathologique, université de Paris, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris-Centre, 20, rue Leblanc, 75015 Paris, France
| | - Sophie Ferlicot
- Réseau national de référence pour cancers rares de l'adulte PREDIR (« Maladie de von Hippel-Lindau et prédispositions héréditaires au cancer rénal ») labellisée par l'Institut national du cancer, université Paris Saclay, Assistance publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Service d'anatomie pathologique des hôpitaux universitaires Paris Sud, université Paris Saclay, hôpital de Bicêtre, Assistance publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Génétique oncologique EPHE, PSL Université, UMR 9019 CNRS, université Paris-Saclay, institut Gustave-Roussy, Villejuif, France
| | - Stéphane Richard
- Réseau national de référence pour cancers rares de l'adulte PREDIR (« Maladie de von Hippel-Lindau et prédispositions héréditaires au cancer rénal ») labellisée par l'Institut national du cancer, université Paris Saclay, Assistance publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France; Génétique oncologique EPHE, PSL Université, UMR 9019 CNRS, université Paris-Saclay, institut Gustave-Roussy, Villejuif, France
| |
Collapse
|