1
|
Wang JJ, Huang RR, Cone BD, Kang SHL, Setoodeh R, Sisk AE, Sajed DP, Shuch BM, Sowalsky AG, Ye H. ELOC-Mutated Renal Cell Carcinoma is a Rare Indolent Tumor with Distinctive Genomic Characteristics. Mod Pathol 2025:100777. [PMID: 40246078 DOI: 10.1016/j.modpat.2025.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/19/2025]
Abstract
ELOC-mutated renal cell carcinoma (ELOC-RCC) is a subtype of renal cell carcinoma (RCC) first recognized by the World Health Organization in 2022, molecularly defined by the presence of ELOC mutations and a lack of VHL mutations. Here, we present an institutional series of ELOC-RCC and provide an in-depth genetic comparison to VHL-null ccRCCs (VHL-ccRCCs). Among 1,209 RCCs in our institutional cytogenetics database, we identified 16 candidate cases that were originally classified as clear cell RCC (ccRCC) and exhibited monosomy 8 and intact chromosome 3p. Seven of these 16 candidate cases were diagnosed as ELOC-RCCs based on histomorphology, immunohistochemistry and whole-exome sequencing results. By contrast, ELOC-RCCs had a simpler karyotype of monosomy 8 with few other alterations. Adding six additional ELOC-RCC cases identified from The Cancer Genome Atlas cohort, all 13 ELOC-RCCs exhibited biallelic ELOC inactivation without VHL mutations. These ELOC mutations (Y79C/L/S/N, E92K, A106D, C112Vfs*3) were all located within or close to the VHL-binding domains in ELOC protein. 69% of the ELOC-RCC cases exhibited its characteristic histomorphology. Compared with stage- and grade-matched VHL-ccRCCs, patients with ELOC-RCCs had superior overall survival (hazard ratio [HR]=0.32, 95% CI=0.16-0.61, p=0.02) and progression-free survival (HR=0.16, 95% CI=0.06-0.42, p=0.04). ELOC-RCCs had significantly fewer somatic copy number alterations (SCNA) and greater abundance of the mutational signature SBS1 than VHL-ccRCCs. ELOC-RCCs lacked common chromosomal alterations or gene mutations seen in ccRCC, including PBRM1, SETD2, BAP1, TSC1, TSC2, or mTOR. Most ELOC-RCCs had linear phylogenetic trees with clonal and truncal ELOC mutations, while additional alterations to ELOC or 8q losses occurred as a subclonal event. While 11 of 13 ELOC-RCCs were confined to the kidney, two ELOC-RCCs were high-stage and exhibited a large solid alveolar pattern, tumor necrosis, more SCNAs, and an additional monoallelic VHL copy loss. Taken together, ELOC-RCCs exhibit distinctive genomic features and indolent behavior in general, supporting it as an independent diagnostic entity.
Collapse
Affiliation(s)
- Jasmine J Wang
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rong Rong Huang
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Brian D Cone
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Sung-Hae L Kang
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Reza Setoodeh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Anthony E Sisk
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Dipti P Sajed
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Brian M Shuch
- Department of Urology, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Adam G Sowalsky
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD, USA.
| | - Huihui Ye
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Gupta S, Cheville JC. Renal Neoplasia: Rare Subtypes and Uncommon Clinical Presentations. Surg Pathol Clin 2025; 18:157-174. [PMID: 39890302 DOI: 10.1016/j.path.2024.09.001] [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] [Indexed: 02/03/2025]
Abstract
Herein, the authors have discussed a series of uncommon familial kidney cancer syndromes (including hyperparathyroidism-jaw tumor syndrome and PTEN hamartoma tumor syndrome), sporadically occurring tumors (BRAF and MTOR pathway-mutated tumors, and juxtaglomerular cell tumors), and uncommon patterns of well-established subtypes of kidney cancer (mucinous tubular spindle cell carcinoma, fumarate hydratase-deficient, and TFE3-rearranged renal cell carcinoma). The rarity of these tumors often leads to diagnostic odysseys for pathologists and patients. Appropriate classification of these rare tumors has implications for screening at-risk family members in the case of hereditary tumor predisposition syndromes, accurate prognostication, and appropriate patient selection for clinical trials.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
3
|
Mansour H, Tran-Dang MA, Walkden M, Boleti E, Barod R, Patki P, Mumtaz F, Tran MGB, Bex A, El Sheikh S. Renal mass biopsy - a practical and clinicopathologically relevant approach to diagnosis. Nat Rev Urol 2025; 22:8-25. [PMID: 38907039 DOI: 10.1038/s41585-024-00897-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 06/23/2024]
Abstract
Advancements in imaging modalities have increased the frequency of renal mass discovery. Imaging has typically been considered sufficient to guide management for a large proportion of these tumours, but renal mass biopsies (RMBs) have an increasing role in determining malignancy and can be a valuable tool for preventing unnecessary surgery in patients with benign tumours. A structured approach should be used to help to navigate the expanding repertoire of renal tumours, many of which are molecularly defined. In terms of tumour subtyping, the pathologist's strategy should focus on stratifying patients into clinically different prognostic groups according to our current knowledge of tumour behaviour, including benign, low-grade or indolent, intermediate malignant or highly aggressive. Crucial pathological features and morphological mimicry of tumours can alter the tumour's prognostic group. Thus, pathologists and urologists can use RMB to select patients with tumours at a reduced risk of progression, which can be safely managed with active surveillance within a tailored imaging schedule, versus tumours for which ablation or surgical intervention is indicated. RMB is also crucial in the oncological setting to distinguish between different high-grade tumours and guide tailored management strategies.
Collapse
Affiliation(s)
- Hussein Mansour
- Research Department of Pathology, UCL Cancer Institute, London, UK
| | - My-Anh Tran-Dang
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
| | - Miles Walkden
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Ekaterini Boleti
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
| | - Ravi Barod
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Prasad Patki
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Faiz Mumtaz
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Maxine G B Tran
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Axel Bex
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK
- UCL Division of Surgery and Interventional Science, Rowland Street, London, UK
| | - Soha El Sheikh
- Research Department of Pathology, UCL Cancer Institute, London, UK.
- Specialist centre for kidney cancer, Royal Free London Hospital, London, UK.
| |
Collapse
|
4
|
Akbari A, Villanueva CR, Hes O, Williamson SR, Kandukuri S, Sharma S, Aditi A, Pivovarcikova K, Argani P, Mohanty SK, Asrani K, Lotan TL. Genetic validation of a TSC2 immunohistochemistry assay in TSC/mTOR-pathway altered renal tumors. Hum Pathol 2024; 154:105693. [PMID: 39571694 DOI: 10.1016/j.humpath.2024.105693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/08/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Pathogenic mutations in the genes associated with tuberous sclerosis complex (TSC)/mTOR pathway are linked to histologically diverse renal cell neoplasms, including eosinophilic solid and cystic renal cell carcinoma (ESC RCC), low grade oncocytic tumor (LOT), eosinophilic vacuolated tumor (EVT), and xanthomatous giant cell renal cell carcinoma (XGC RCC). Here, we validate a TSC2 immunohistochemistry (IHC) assay by comparison to genomic data in these neoplasms. Automated TSC2 IHC was performed on formalin-fixed paraffin embedded (FFPE) tissues from 38 genetically-confirmed TSC/mTOR-associated renal tumors (6 ESCs, 16 EVTs, 13 LOTs, 2 XGC and 1 clear cell RCC) and visually scored in a semi-dichotomous fashion compared to internal control tissue. The positive predictive value (PPV) of TSC2 protein loss for underlying pathogenic mutation in TSC2 was 92% (11/12), while the negative predictive value (NPV) of intact TSC2 by IHC for lack of underlying pathogenic mutation in TSC2 was 81% (21/26). Intact TSC2 by IHC was 95% (21/22) specific for absence of underlying pathogenic TSC2 mutation. All the cases lacking TSC2 mutation with intact TSC2 protein had an underlying mutation in TSC1, MTOR or PIK3CA. Loss of TSC2 was 77% (10/13) sensitive for underlying TSC2 truncation mutations and 33% (1/3) sensitive for underlying TSC2 missense mutations. Overall, 73% (8/11) tumors with TSC2 IHC loss and underlying pathogenic alterations in TSC2 showed heterogeneous protein loss, with rare interspersed positively staining tumor cells. These data support TSC2 IHC as a potentially useful assay for the diagnostic workup of renal tumors suspected to belong to the TSC/mTOR-associated subgroups.
Collapse
Affiliation(s)
- Amir Akbari
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Pathology, University of Southern California Keck School of Medicine, USA.
| | | | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | | | - Shivani Kandukuri
- Department of Pathology, University of Southern California Keck School of Medicine, USA
| | | | | | - Kristyna Pivovarcikova
- Department of Pathology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pedram Argani
- Department of Pathology, Johns Hopkins School of Medicine, USA
| | - Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, OR, India
| | - Kaushal Asrani
- Department of Pathology, Johns Hopkins School of Medicine, USA.
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins School of Medicine, USA; Department of Urology, Johns Hopkins School of Medicine, USA; Department of Oncology, Johns Hopkins School of Medicine, USA.
| |
Collapse
|
5
|
Pezzicoli G, Musci V, Ciciriello F, Salonne F, Cafforio P, Lionetti N, Ragno A, Rizzo M. Genomic profiling and molecular characterization of non-clear cell renal cell carcinoma: a narrative review from a clinical perspective. Ther Adv Med Oncol 2024; 16:17588359241298500. [PMID: 39563719 PMCID: PMC11574901 DOI: 10.1177/17588359241298500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
While the clear-cell renal cell carcinoma (ccRCC) treatment has undergone several paradigm shifts in recent years, the non-clear cell renal cell carcinoma (nccRCC) therapeutic approach has yet to be extensively investigated and improved. The WHO 2022 classification of renal neoplasms redefined the most common nccRCC subtypes (papillary and chromophobe RCC) and introduced the molecularly defined RCC class, which is a first step in the direction of better molecular profiling of nccRCC. We reviewed the literature data on known genomic alterations of clinical interest in nccRCC and discussed their potential role in guiding therapeutic choices in each nccRCC entity. Among the alterations discussed, we focused on the ones that could be treated with already available drugs, such as MET-driven papillary RCC, mechanistic target of rapamycin altered chromophobe RCC, anaplastic lymphoma kinase-rearranged RCC, and fumarate-hydratase deficient RCC. Furthermore, we focused on the currently ongoing clinical trials and further evidence for all the other entities, such as SMARCB1-deficient RCC, TFE3 and transcription factorEB (TFEB)-altered RCC, and Elongin C (ELOC)-mutated RCC. The vast heterogeneity of nccRCC does not allow a one-size-fits-all solution; therefore, molecular characterization is the path toward effective therapies and fully personalized medicine for these entities.
Collapse
Affiliation(s)
- Gaetano Pezzicoli
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Vittoria Musci
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Federica Ciciriello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Francesco Salonne
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Paola Cafforio
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Nicoletta Lionetti
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Anna Ragno
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Consorziale, Policlinico di Bari, Bari, Italy
| | - Mimma Rizzo
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Consorziale, Policlinico di Bari, piazza G. Cesare 11, Bari 70124, Italy
| |
Collapse
|
6
|
Gupta S, McCarthy MR, Tjota MY, Antic T, Cheville JC. Metastatic renal cell carcinoma with fibromyomatous stroma associated with tuberous sclerosis or MTOR, TSC1/TSC2-Mutations: A Series of 4 cases and a review of the literature. Hum Pathol 2024; 153:105680. [PMID: 39522702 DOI: 10.1016/j.humpath.2024.105680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Renal cell carcinoma with fibromyomatous stroma (RCCfms) are characterized by a constellation of morphologic findings that include elongated tubules lined by cells with clear to pale eosinophilic cytoplasm and intersecting bands of smooth muscle stroma. Consistent immunohistochemistry findings in RCCfms include diffuse positivity for carbonic anhydrase 9 and variable expression of keratin 7. Molecular profiling of such tumors show either pathogenic alterations of the ELOC (TCEB1) gene, or alterations of MTOR, TSC1, and TSC2. MTOR, TSC1/TSC2-altered RCCfms (M/TSC-RCCfms) has been reported both in the sporadic setting and in association with tuberous sclerosis complex (TSC). The importance of accurate diagnosis of M/TSC-RCCfms includes prompting germline testing in the appropriate clinical context. In addition, it can lead to patient management strategies that are focused on the preservation of renal function, as TSC patients often have multifocal and bilateral disease. As diagnostic criteria for M/TSC-RCCfms have only been recently established, additional data are needed to understand the natural history of this disease. Herein, we report 6 patients with metastatic M/TSC-RCCfms, including four patients from our institutional archives (four males, aged 36-58 years at nephrectomy), and two additional cases reported in the literature. Five patients had TSC, and the sixth had an MTOR-altered RCCfms. The majority of patients (5/6, 83%) presented with regional lymph node involvement and one patient developed metastases to the lung. All patients were alive at last follow up (median follow-up of 85 months). Our report is intended to raise awareness regarding rare instances of metastatic behavior for M/TSC-RCCfms.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Michael R McCarthy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Melissa Y Tjota
- Department of Pathology, The University of Chicago, IL, 60637, USA.
| | - Tatjana Antic
- Department of Pathology, The University of Chicago, IL, 60637, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
7
|
Palathingal Bava E, Gupta N, Alruwaii FI, Nelson R, Al-Obaidy KI. Recurrent MTOR Mutations in Renal Cell Carcinoma With Fibromyomatous Stroma: A Report of 2 Tumors. Int J Surg Pathol 2024; 32:1409-1414. [PMID: 38311893 DOI: 10.1177/10668969241228295] [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] [Indexed: 02/06/2024]
Abstract
Renal cell carcinoma with fibromyomatous stroma, recognized as a provisional entity in the current 2022 World Health Organization classification of renal neoplasms, is rare. Recent evidence suggests recurrent alterations in the mTOR pathway, supporting its recognition as a distinct entity. Herein, we report 2 renal cell carcinomas with fibromyomatous stroma with MTOR mutations occurring in 62- and 72-year-old women and review the literature to support its recognition as a distinct entity, focusing on the characteristic morphology, immunohistochemical staining patterns as well as genetic alterations.
Collapse
Affiliation(s)
- Ejas Palathingal Bava
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
| | - Nilesh Gupta
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Fatimah I Alruwaii
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
| | - Ryan Nelson
- Department of Urology, Henry Ford Health, Detroit, MI, USA
| | - Khaleel I Al-Obaidy
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
- Department of Urology, Henry Ford Health, Detroit, MI, USA
| |
Collapse
|
8
|
Pacyna CN, Anandapadamanaban M, Loudon KW, Hay IM, Perisic O, Li R, Byrne M, Allen L, Roberts K, Hooks Y, Warren AY, Stewart GD, Clatworthy MR, Teichmann SA, Behjati S, Campbell PJ, Williams RL, Mitchell TJ. Multifocal, multiphenotypic tumours arising from an MTOR mutation acquired in early embryogenesis. Oncogene 2024; 43:3268-3276. [PMID: 39271965 PMCID: PMC11518995 DOI: 10.1038/s41388-024-03137-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024]
Abstract
Embryogenesis is a vulnerable time. Mutations in developmental cells can result in the wide dissemination of cells predisposed to disease within mature organs. We characterised the evolutionary history of four synchronous renal tumours from a 14-year-old girl using whole genome sequencing alongside single cell and bulk transcriptomic sequencing. Phylogenetic reconstruction timed the origin of all tumours to a multipotent embryonic cell committed to the right kidney, around 4 weeks post-conception. Biochemical and structural analysis of their shared MTOR mutation, absent from normal tissues, demonstrates enhanced protein flexibility, enabling a FAT domain hinge to dramatically increase activity of mTORC1 and mTORC2. Developmental mutations, not usually detected in traditional genetic screening, have vital clinical importance in guiding prognosis, targeted treatment, and family screening decisions for paediatric tumours.
Collapse
Affiliation(s)
- Clarissa N Pacyna
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Kevin W Loudon
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Iain M Hay
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Olga Perisic
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Ruoyan Li
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Matthew Byrne
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Laura Allen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Kirsty Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Yvette Hooks
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Anne Y Warren
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Grant D Stewart
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Menna R Clatworthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
- Department of Paediatrics, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Peter J Campbell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | | | - Thomas J Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK.
- Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK.
- Department of Surgery, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
9
|
Alruwaii ZI, Williamson SR, Al-Obaidy KI. Mechanistic Target of Rapamycin Kinase is a Common Convergent Pathway to Renal Neoplasia: A Contemporary Review. Int J Surg Pathol 2024; 32:1095-1108. [PMID: 38258297 DOI: 10.1177/10668969231219653] [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] [Indexed: 01/24/2024]
Abstract
Mechanistic target of rapamycin kinase (mTOR) is a member of the phosphatidylinositol-3-hydroxide kinase (PI3 K)-related protein kinase family that functions as a central regulator of cell growth, metabolism, proliferation, and survival. The role of the TSC-mTOR signaling pathway in kidney tumors has been implicated in some hamartoma syndromes; however, with the advent and wide utilization of molecular studies, a growing number of kidney tumors have been linked to somatic or germline mutations involving genes that encode for this pathway, including eosinophilic solid and cystic renal cell carcinoma, low-grade oncocytic tumor, eosinophilic vacuolated tumor, renal cell carcinoma with fibromyomatous stroma and angiomyolipoma, among others. Herein, we review the contemporary developments of mTOR pathway-related renal neoplasia, focusing on the clinicopathologic features of the tumor entities.
Collapse
Affiliation(s)
- Zainab I Alruwaii
- Department of Pathology and Laboratory Medicine, King Fahad Specialist Hospital, Dammam, KSA
| | - Sean R Williamson
- Pathology and Laboratory Medicine Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - Khaleel I Al-Obaidy
- Department of Pathology and Laboratory Medicine, Henry Ford Health, Detroit, MI, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
10
|
Liu Y, Zhou L, Xu H, Gu Y, Dong L, Yang X, Wang C. Mutated ASXL1 upregulates mTOR expression in renal cell carcinoma with fibromyomatous stroma. Virchows Arch 2024; 485:379-382. [PMID: 38102390 DOI: 10.1007/s00428-023-03667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 12/17/2023]
Abstract
Renal cell carcinoma with fibromyomatous stroma (RCC FMS), defined as an "emerging entity" in the 2016 WHO classification and recommended to be a novel entity by GUPS, is represented by tumor cells with clear to mildly eosinophilic cytoplasm displaying elongated and branching tubules and papillae. A fibromyomatous stroma could be observed in these tumors. These tumors are immunopositive for CK7 and featured by ELOC and/or TSC/mTOR gene mutations. In the 2022 WHO classification, ELOC mutated RCC is classified as a molecularly defined RCCs as an individual renal entity. However, there are limited descriptions of TSC/mTOR alterations in RCC FMS. Herein, we reported a case of 28-year-old woman with RCC FMS with intact ELOC and TSC/mTOR genes but ASXL1 mutation. The tumor cells were positive for mTOR expression. This case may indicate that altered mTOR expression, but not limited to mutated TSC/mTOR gene, that participates in the pathogenesis of RCC FMS.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China
| | - Luting Zhou
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China
| | - Haimin Xu
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China
| | - Yijin Gu
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China
| | - Lei Dong
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China
| | - Xiaoqun Yang
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China.
| | - Chaofu Wang
- Department of Pathology, Shanghai Jiaotong University Medical School Affiliated Ruijin Hospital, Number 197, Ruijin Er Road, Huangpu District, Shanghai, China.
| |
Collapse
|
11
|
Kanakaraj J, Chang J, Hampton LJ, Smith SC. The New WHO Category of "Molecularly Defined Renal Carcinomas": Clinical and Diagnostic Features and Management Implications. Urol Oncol 2024; 42:211-219. [PMID: 38519377 DOI: 10.1016/j.urolonc.2024.02.003] [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: 08/22/2023] [Revised: 01/27/2024] [Accepted: 02/12/2024] [Indexed: 03/24/2024]
Abstract
The evolution of classification of renal tumors has been impacted since the turn of the millennium by rapid progress in histopathology, immunohistochemistry, and molecular genetics. Together, these features have enabled firm recognition of specific, classic types of renal cell carcinomas, such as clear cell renal cell carcinoma, that in current practice trigger histologic-type specific management and treatment protocols. Now, the fifth Edition World Health Classification's new category of "Molecularly defined renal carcinomas" changes the paradigm, defining a total of seven entities based specifically on their fundamental molecular underpinnings. These tumors, which include TFE3-rearranged, TFEB-altered, ELOC-mutated, fumarate hydratase-deficient, succinate dehydrogenase-deficient, ALK-rearranged, and SMARCB1-deficient renal medullary carcinoma, encompass a wide clinical and histopathologic phenotypic spectrum of tumors. Already, important management aspects are apparent for several of these entities, while emerging therapeutic angles are coming into view. A brief, clinically-oriented introduction of the entities in this new category, focusing on relevant diagnostic, molecular, and management aspects, is the subject of this review.
Collapse
Affiliation(s)
- Jonathan Kanakaraj
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Justin Chang
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Lance J Hampton
- Division of Urology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Pathology, Richmond Veterans Affairs Medical Center, Richmond, VA; VCU Massey Comprehensive Cancer Center, Richmond, VA
| | - Steven Christopher Smith
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA; Division of Urology, Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA; Department of Pathology, Richmond Veterans Affairs Medical Center, Richmond, VA; VCU Massey Comprehensive Cancer Center, Richmond, VA.
| |
Collapse
|
12
|
Wen N, Li X, Lu J, Pan L, Yang P, Zhang Y, Chen K, Cao Y. Quantification of eosinophilic area and its potential molecular feature in clear cell renal cell carcinoma. Jpn J Clin Oncol 2024; 54:689-698. [PMID: 38366664 DOI: 10.1093/jjco/hyae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/27/2024] [Indexed: 02/18/2024] Open
Abstract
OBJECTIVE Previous studies have acknowledged the presence of eosinophilic cytoplasm in clear cell renal cell carcinoma, yet the precise quantification method and potential molecular attributes in clear cell renal cell carcinoma remain elusive. This study endeavours to precisely quantify the eosinophilic attribute and probe into the molecular mechanisms governing its presence in clear cell renal cell carcinoma. METHODS Data from cohorts of clear cell renal cell carcinoma patients who underwent nephrectomy, comprising The Cancer Genome Atlas cohort (n = 475) and Sun Yat-sen University Cancer Center cohort (n = 480), were aggregated to assess the eosinophilic attribute. Additionally, Omics data from Clinical Proteomic Tumor Analysis Consortium (CPTAC) (n = 58) were leveraged to explore the potential molecular features associated with eosinophilic clear cell renal cell carcinoma. Employing receiver operating characteristic curve analysis, the proportion of tumour cells with eosinophilic cytoplasm was determined, leading to the classification of each cohort into distinct groups: a clear group (<5%) and an eosinophilic group (≥5%). RESULTS In both cohorts, the eosinophilic feature consistently correlated with higher International Society of Urological Pathology (ISUP) grade, elevated tumor stage, and the presence of necrosis. Furthermore, the Kaplan-Meier method demonstrated that patients in the eosinophilic group exhibited shorter overall survival or disease-free survival compared with those in the clear group, a pattern reaffirmed in various stratified survival analyses. Intriguingly, within The Cancer Genome Atlas cohort, the pathological characterization of cell cytoplasm (eosinophilic vs. clear) emerged as an independent risk factor for overall survival (hazard ratio = 2.507 [95% confidence interval: 1.328-4.733], P = 0.005) or disease-free survival (hazard ratio = 1.730 [95% confidence interval: 1.062-2.818], P = 0.028) via Cox regression analysis. Moreover, multi-Omics data unveiled frequent BAP1 mutations and down-regulation of Erythroblast Transformation-Specific-Related Gene associated with the eosinophilic feature in clear cell renal cell carcinoma. Additionally, patients with low expression of Erythroblast Transformation-Specific-Related Gene showed worse overall survival (P < 0.001). CONCLUSIONS The quantification of the eosinophilic feature serves as a robust predictor of clinical prognosis in clear cell renal cell carcinoma. Furthermore, the manifestation of this feature may be linked to BAP1 mutations and the down-regulation of Erythroblast Transformation-Specific-Related Gene in clear cell renal cell carcinoma. Significantly, the expression levels of Erythroblast Transformation-Specific-Related Gene manifest as an exemplary prognostic marker, providing exceptional predictive accuracy for the clinical prognosis in clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Nengqiao Wen
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Xiaomin Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jiangli Lu
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Lu Pan
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ping Yang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yijun Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Keming Chen
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yun Cao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
13
|
Pei J, Yan Y, Jayaraman S, Rajagopal P, Natarajan PM, Umapathy VR, Gopathy S, Roy JR, Sadagopan JC, Thalamati D, Palanisamy CP, Mironescu M. A review on advancements in the application of starch-based nanomaterials in biomedicine: Precision drug delivery and cancer therapy. Int J Biol Macromol 2024; 265:130746. [PMID: 38467219 DOI: 10.1016/j.ijbiomac.2024.130746] [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: 10/07/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
The burgeoning field of starch-based nanomaterials in biomedical applications has perceived notable progressions, with a particular emphasis on their pivotal role in precision drug delivery and the inhibition of tumor growth. The complicated challenges in current biomedical research require innovative approaches for improved therapeutic outcomes, prompting an exploration into the possible of starch-based nanomaterials. The conceptualization of this review emerged from recognizing the need for a comprehensive examination of the structural attributes, versatile properties, and mechanisms underlying the efficiency of starch-based nanomaterials in inhibiting tumor growth and enabling targeted drug delivery. This review delineates the substantial growth in utilizing starch-based nanomaterials, elucidating their small size, high surface-volume ratio, and biocompatibility, predominantly emphasizing their possible to actively recognize cancer cells, deliver anticancer drugs, and combat tumors efficiently. The investigation of these nanomaterials encompasses to improving biocompatibility and targeting specific tissues, thereby contributing to the evolving landscape of precision medicine. The review accomplishes by highlighting the auspicious strategies and modern developments in the field, envisioning a future where starch-based nanomaterials play a transformative role in molecular nanomaterials, evolving biomedical sciences. The translation of these advancements into clinical applications holds the potential to revolutionize targeted drug delivery and expand therapeutic outcomes in the realm of precision medicine.
Collapse
Affiliation(s)
- JinJin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - Yuqiang Yan
- Department of anaesthesia, Xi'an Central Hospital, No. 161, West 5th Road, Xincheng District, Xi'an 710003, China
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai-600 095, India
| | - Prabhu Manickam Natarajan
- Department of Clinical Sciences, Center of Medical and Bio-allied Health Sciences and Research, College of Dentistry, Ajman University, Ajman, United Arab Emirates
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Thai Moogambigai Dental College and Hospital, Chennai-600107, India
| | - Sridevi Gopathy
- Department of Physiology, SRM Dental College, Ramapuram campus, Chennai 600089, India
| | - Jeane Rebecca Roy
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600 073, India
| | - Janaki Coimbatore Sadagopan
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600 073, India
| | | | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Monica Mironescu
- Faculty of Agricultural Sciences Food Industry and Environmental Protection, Lucian Blaga University of Sibiu, Sibiu 550024, Romania.
| |
Collapse
|
14
|
Collins K, Kocemba J, Saad M, Acosta AM, Ulbright TM, Argani P, Williamson SR. Smooth muscle and adenoma-like renal tumour (SMART): another example suggesting distinction from mixed epithelial and stromal tumour. Histopathology 2024; 84:903-905. [PMID: 38114288 DOI: 10.1111/his.15119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/23/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Affiliation(s)
- Katrina Collins
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua Kocemba
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mohammed Saad
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andres M Acosta
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas M Ulbright
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
15
|
Siegmund SE, Al-Obaidy KI, Tsai HK, Idrees MT, Akgul M, Acosta AM, Hirsch MS. Concordance of MTOR Pathway Mutations and the Diagnosis of Renal Low-Grade Oncocytic Tumor (LOT). Int J Surg Pathol 2024; 32:316-330. [PMID: 37357748 DOI: 10.1177/10668969231178032] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The differential diagnosis for oncocytic renal tumors spans the spectrum from benign entities to more aggressive renal cell carcinomas (RCC). Recent work has characterized a provisional renal oncocytic neoplasm, namely the low-grade oncocytic tumor (LOT), which demonstrates overlapping morphologic features with oncocytoma and chromophobe RCC, but also has a unique immunoprofile (ie, diffusely positive for KRT7, negative for KIT) and a high rate (80% to 100%) of mTOR pathway gene alterations. Given the diagnostic overlap among oncocytic tumors, we looked for concordance between mTOR pathway mutations and LOT. Thirty low-grade renal oncocytic neoplasms underwent histologic review and immunohistochemistry for KRT7 and KIT. Tumors were classified as "determinate" (eg, LOT) for tumors with solid, nested or vaguely tubular growth and diffuse KRT7 staining and negative KIT, or "indeterminate" if the morphology and/or immunostains did not fully support a definitive LOT diagnosis. Next-generation sequencing was performed without any knowledge of the diagnoses, and identified mTOR pathway mutations in 80% (12/15) of the determinate tumors, compared with 7% (1/15) in the indeterminate group. One determinate tumor was reclassified as papillary RCC (MTOR mutation negative) and 6 indeterminate tumors were confirmed to be oncocytoma (N = 4), clear cell RCC or papillary RCC with reverse polarity, respectively. Overall, integration of morphology, immunohistochemistry, and molecular data enabled a final definitive diagnosis for 70% of tumors (21 of the total 30), with a high concordance (93%) for LOT specifically in the determinate group; the remaining 9 tumors (30%) were classified as renal oncocytic neoplasm, not otherwise specified.
Collapse
Affiliation(s)
- Stephanie E Siegmund
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Khaleel I Al-Obaidy
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Harrison K Tsai
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Advanced Molecular Diagnostics, Brigham and Women's Hospital, Boston, MA, USA
| | - Muhammad T Idrees
- Department of Pathology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mahmut Akgul
- Department of Pathology, Albany Medical Center, Albany, NY, USA
| | - Andres M Acosta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Goswami PR, Singh G, Patel T, Dave R. The WHO 2022 Classification of Renal Neoplasms (5th Edition): Salient Updates. Cureus 2024; 16:e58470. [PMID: 38765391 PMCID: PMC11100973 DOI: 10.7759/cureus.58470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2024] [Indexed: 05/22/2024] Open
Abstract
The first categorization for renal tumours was made by the WHO in 1981 and included only renal cell carcinoma (RCC). After that, classification was continuously altered over five decades. The WHO 2022 Classification of Urinary and Male Genital Tumours 2022 (5th edition) is molecular-driven and contains major revisions compared to the earlier classification from 2016. This revised edition divided renal tumours into four major broad categories: clear cell renal tumours, papillary renal cell tumours, oncocytic and chromophobe renal tumours, and collecting duct tumours. 'Other renal tumours' and 'molecularly defined renal carcinomas' are two other categories that were also included. Transcription factor binding to IGHM enhancer 3 (TFE3)-rearranged, TFEB-altered, elongin C (ELOC)-mutated (formerly TCEB1)-mutated, fumarate hydratase (FH)-deficient, succinate dehydrogenase (SDH)-deficient, anaplastic lymphoma kinase (ALK)-rearranged, and SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1)-deficient renal cell carcinomas are molecularly defined entities. Eosinophilic vacuolated tumours and low-grade oncocytic tumours are classified as emerging entities. Molecularly characterized renal tumours include those with SMARCB1 deficiencies, TFE3 rearrangements, TFEB alterations, ALK rearrangements, ELOC mutations, etc. Thyroid-like follicular carcinoma, eosinophilic vacuolated tumour, and low-grade oncocytic tumour are a few emerging entities of renal tumours. Improved therapy targets for each kidney tumour can be achieved using immunohistochemistry (IHC) and molecular definition updates. This study aims to highlight new developments in the WHO 2022 categorization of renal tumours with regard to diagnostic, morphological, molecular, IHC, clinical, and prognostic updates.
Collapse
Affiliation(s)
- Parth R Goswami
- Pathology, All India Institute of Medical Sciences (AIIMS), Rajkot, Gujarat, IND
| | - Gyanendra Singh
- Pathology, All India Institute of Medical Sciences (AIIMS), Rajkot, IND
| | - Tarang Patel
- Pathology, All India Institute of Medical Sciences (AIIMS), Rajkot, IND
| | - Rushang Dave
- Pathology, Shantabaa Medical College and General Hospital, Amreli, IND
| |
Collapse
|
17
|
Shah RB, Mehra R. Renal Cell Carcinoma Associated With TSC/MTOR Genomic Alterations: An Update on its Expanding Spectrum and an Approach to Clinicopathologic Work-up. Adv Anat Pathol 2024; 31:105-117. [PMID: 37899532 DOI: 10.1097/pap.0000000000000419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
Renal cell carcinoma (RCC) with tuberous sclerosis complex (TSC)/mammalian target of rapamycin (MTOR) pathway-related genomic alterations have been classically described in hereditary TSC syndrome setting involving germline mutations, whereby cells with a bi-allelic inactivation of genes originate tumors in a classic tumor-suppressor "two-hit" Knudson paradigm. Initial studies of TSC-associated RCC categorized tumors into 3 broad heterogeneous morphologic groups: RCC with smooth muscle stroma, chromophobe-like, and eosinophilic-macrocytic. Recently, a similar morphologic spectrum has been increasingly recognized in novel and emerging entities characterized by somatic mutations in the TSC1/2 and MTOR in patients who do not suffer from the TSC. Correct recognition of RCC with TSC / MTOR mutations is critical for accurate prognostication because such tumors with aggressive behavior have the potential to be tailored to mTOR inhibitors. Whether TSC/MTOR mutated renal epithelial neoplasms represent a distinct molecular class has been confounded by the fact that TSC1/2 , and the gene encoding the downstream protein MTOR, are mutated secondarily in ∼5% of the more common subtypes of RCC, including the commonest subtype of clear cell RCC. This review summarizes the expanding morphologic spectrum of renal tumors with TSC/mTOR pathway alterations, specifically for sporadically occurring tumors where these genomic alterations likely are primary pathologic events. Finally, a practical surgical pathology approach to handling these tumors, and a conceptual framework of renal epithelial tumors with TSC/MTOR mutations as a "family of tumors", is presented.
Collapse
Affiliation(s)
- Rajal B Shah
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX
| | - Rohit Mehra
- Department of Pathology and Michigan Center for Translational Pathology, University of Michigan School of Medicine, Ann Arbor, MI
| |
Collapse
|
18
|
Kojima F, Matsuzaki I, Musangile FY, Kinoshita Y, Otani T, Abe K, Asai A, Kohjimoto Y, Kondo T, Hara I, Murata SI. Clinicopathological and molecular features of renal cell carcinomas with haemangioblastoma-like features distinct from clear cell renal cell carcinoma. Histopathology 2024; 84:539-549. [PMID: 37988260 DOI: 10.1111/his.15098] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/23/2023]
Abstract
AIMS Haemangioblastomas arise in the central nervous system. Rarely, haemangioblastomas may develop in extra-neural sites, such as the kidneys. A few reported cases of renal cell carcinomas (RCCs) with haemangioblastoma-like features have exhibited both clear cell renal cell carcinoma (CCRCC)- and haemangioblastoma-like components. The clinicopathological and molecular characteristics of RCCs with haemangioblastoma-like features were analysed, focusing on VHL alterations, in comparison with CCRCCs partially resembling haemangioblastoma. METHODS AND RESULTS Four RCCs with haemangioblastoma-like features and five CCRCCs partially resembling haemangioblastoma were included. The RCCs with haemangioblastoma-like features were indolent and lacked adverse prognostic factors. All RCCs with haemangioblastoma-like features had a well-circumscribed appearance and a thick fibromuscular capsule, with fibromuscular bundles extending into the tumour to varying degrees in the three tumours. Each RCC with haemangioblastoma-like features exhibited CCRCC-like areas with indistinct tubular structures and foci of haemangioblastoma-like areas, in which vessels and short spindle cells overwhelmed tumour cells. Whereas haemangioblastoma-like areas in the CCRCCs partially resembling haemangioblastoma exhibited sparse vessels and spindle cells and distinct clear cells. The RCCs with haemangioblastoma-like features exhibited a unique immunohistochemical profile, with positive staining for inhibin-α, S100, carbonic-anhydrase-9, keratin7, and high molecular weight keratin and negative staining for (alpha-methylacyl-CoA racemase) AMACR. RCC with haemangioblastoma-like features did not display any VHL alterations, including VHL mutation, 3p LOH, and methylation of the VHL promoter region, and the two tumours harboured a likely oncogenic missense variant of MTOR (c.7280T>G). CONCLUSION The histopathological, immunohistochemical, and molecular findings suggest that RCC with haemangioblastoma-like features is a distinct entity from CCRCC.
Collapse
Affiliation(s)
- Fumiyoshi Kojima
- Department of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | - Ibu Matsuzaki
- Department of Human Pathology, Wakayama Medical University, Wakayama, Japan
| | | | - Yuichi Kinoshita
- Department of Clinical Laboratory, Wakayama Medical University, Wakayama, Japan
| | - Toshinori Otani
- Department of Pathology, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Kuniko Abe
- Department of Diagnostic Pathology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Akihiro Asai
- Department of Urology, Japanese Red Cross Nagasaki Genbaku Hospital, Nagasaki, Japan
| | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Tetsuo Kondo
- Department of Human Pathology, University of Yamanashi, Yamanashi, Japan
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Shin-Ichi Murata
- Department of Human Pathology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
19
|
Machacek ME, Wu CL, Cornejo KM. Pathology of hereditary renal cell carcinoma syndromes: Tuberous sclerosis complex (TSC). Semin Diagn Pathol 2024; 41:8-19. [PMID: 37993384 DOI: 10.1053/j.semdp.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 11/24/2023]
Abstract
Tuberous sclerosis complex (TSC) is an autosomal dominant genetic disease characterized by hamartomatous tumors involving multiple organs such as the brain, skin, heart, lung and kidney. TSC is caused by inactivating mutations in TSC1/TSC2, which encodes hamartin and tuberin, respectively, and forms a complex that regulates mechanistic target of rapamycin complex 1 (mTORC1), resulting in cell overgrowth and oncogenesis. Since a leading cause of morbidity and mortality in TSC relates to chronic kidney disease and the ability to preserve renal function, this review describes the important pathologic findings in TSC-associated renal neoplasms and their correlating sporadic counterparts. The most common renal tumor in TSC patients are AMLs, followed by a heterogeneous spectrum of renal epithelial tumors, which may provide clues to establishing a diagnosis of TSC.
Collapse
Affiliation(s)
- Miranda E Machacek
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Kristine M Cornejo
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
20
|
Alchoueiry M, Cornejo K, Henske EP. Kidney cancer: Links between hereditary syndromes and sporadic tumorigenesis. Semin Diagn Pathol 2024; 41:1-7. [PMID: 38008653 DOI: 10.1053/j.semdp.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023]
Abstract
Multiple hereditary syndromes predispose to kidney cancer, including Von Hippel-Lindau syndrome, BAP1-Tumor Predisposition Syndrome, Hereditary Papillary Renal Cell Carcinoma, Tuberous Sclerosis Complex, Birt-Hogg-Dubé syndrome, Hereditary Paraganglioma-Pheochromocytoma Syndrome, Fumarate Hydratase Tumor Predisposition Syndrome, and Cowden syndrome. In some cases, mutations in the genes that cause hereditary kidney cancer are tightly linked to similar histologic features in sporadic RCC. For example, clear cell RCC occurs in the hereditary syndrome VHL, and sporadic ccRCC usually has inactivation of the VHL gene. In contrast, mutations in FLCN, the causative gene for Birt-Hogg-Dube syndrome, are rarely found in sporadic RCC. Here, we focus on the genes and pathways that link hereditary and sporadic RCC.
Collapse
Affiliation(s)
- Michel Alchoueiry
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristine Cornejo
- Pathology Department, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth P Henske
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Caliò A, Marletta S, Settanni G, Rizzo M, Gobbo S, Pedron S, Stefanizzi L, Munari E, Brunelli M, Marcolini L, Pesci A, Fratoni S, Pierconti F, Raspollini MR, Marchetti A, Doglioni C, Amin MB, Porta C, Martignoni G. mTOR eosinophilic renal cell carcinoma: a distinctive tumor characterized by mTOR mutation, loss of chromosome 1, cathepsin-K expression, and response to target therapy. Virchows Arch 2023; 483:821-833. [PMID: 37938323 PMCID: PMC10700445 DOI: 10.1007/s00428-023-03688-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/17/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023]
Abstract
In the spectrum of oncocytic renal neoplasms, a subset of tumors with high-grade-appearing histologic features harboring pathogenic mutations in mammalian target of rapamycin (mTOR) and hitherto clinical indolent behavior has been described. Three cases (2F,1 M) with histologically documented metastases (lymph node, skull, and liver) were retrieved and extensively investigated by immunohistochemistry, FISH, and next-generation sequencing. Tumors were composed of eosinophilic cells with prominent nucleoli (G3 by ISUP/WHO) arranged in solid to nested architecture. Additionally, there were larger cells with perinuclear cytoplasmic shrinkage and sparse basophilic Nissl-like granules, superficially resembling the so-called spider cells of cardiac rhabdomyomas. The renal tumors, including the skull and liver metastases, showed immunoexpression PAX8, CK8-18, and cathepsin-K, and negativity for vimentin. NGS identified mTOR genetic alterations in the three cases, including the skull and liver metastases. One patient was then treated with Everolimus (mTOR inhibitors) with clinical response (metastatic tumor shrinkage). We present a distinct renal tumor characterized by high-grade eosinophilic cells, cathepsin-K immunohistochemical expression, and harboring mTOR gene mutations demonstrating a malignant potential and showing responsiveness to mTOR inhibitors.
Collapse
Affiliation(s)
- Anna Caliò
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Stefano Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Giulio Settanni
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Mimma Rizzo
- Division of Medical Oncology, A.O.U. Consorziale Policlinico Di Bari, Bari, Italy
| | - Stefano Gobbo
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Serena Pedron
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | | | - Enrico Munari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Brunelli
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy
| | - Lisa Marcolini
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Anna Pesci
- Department of Pathology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| | - Stefano Fratoni
- Division of Anatomic Pathology, S. Eugenio Hospital, Rome, Italy
| | - Francesco Pierconti
- Division of Anatomic Pathology and Histology, Foundation "A. Gemelli" University Hospital, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Rosaria Raspollini
- Histopathology and Molecular Diagnostics, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Antonio Marchetti
- Division of Anatomic Pathology and Histology, Ospedale Clinicizzato "SS. Annunziata" Università Di Chieti, Chieti, Italy
| | | | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science, Memphis, TN, USA
- Department of Urology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari "A. Moro, Bari, Italy
| | - Guido Martignoni
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Largo L. Scuro 10, 37134, Verona, Italy.
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| |
Collapse
|
22
|
Li H, Argani P, Halper-Stromberg E, Lotan TL, Merino MJ, Reuter VE, Matoso A. Positive GPNMB Immunostaining Differentiates Renal Cell Carcinoma With Fibromyomatous Stroma Associated With TSC1/2/MTOR Alterations From Others. Am J Surg Pathol 2023; 47:1267-1273. [PMID: 37661807 PMCID: PMC10592185 DOI: 10.1097/pas.0000000000002117] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Renal cell carcinoma with fibromyomatous stroma (RCCFMS) include ELOC/TCEB1 -mutated renal cell carcinoma (RCC) and those with TSC1/2 / MTOR alterations. Besides morphologic similarity, most of these tumors is known to be diffusely positive for carbonic anhydrase IX and cytokeratin 7 by immunohistochemistry. We previously showed strong and diffuse expression of GPNMB (glycoprotein nonmetastatic B) in translocation RCC and eosinophilic renal neoplasms with known TSC1/2/MTOR alterations. We retrospectively identified molecularly confirmed cases of TCEB1/ELOC -mutated RCC (7 tumors from 7 patients), and RCCFMS with alterations in TSC1/2/MTOR (6 tumors from 5 patients, 1 patient with tuberous sclerosis syndrome). In addition, we included 7 clear cell papillary renal cell tumors (CCPRCTs) and 8 clear cell RCC, as they can also present morphologic overlap with RCCFMS. Morphologically, RCCs with TSC1/2/MTOR alterations and those with TCEB1/ELOC mutations were indistinguishable and characterized by papillary, nested, or tubular architecture, with tumor cells with clear cytoplasm and low nuclear grade. By immunohistochemistry, cytokeratin 7 was positive in 5/7 (71%) of TCEB1/ELOC -mutated RCCs, 6/6 (100%) of RCCs with TSC1/2/mTOR alterations, and 7/7 (100%) of CCPRCTs ( P =not significant). Carbonic anhydrase IX was positive in 7/7 TCEB1/ELOC -mutated RCCs, 6/6 (100%) of RCCs with TSC1/2/MTOR alterations, and 7/7 (100%) of CCPRCTs ( P =NS). GPNMB was strongly and diffusely positive in all tumors with TSC1/2/MTOR alterations (6/6), while negative in all TCEB1/ELOC -mutated RCCs (0/6), or CCPRCTs (0/7) ( P =0.002). Two of 8 clear cell RCC showed focal weak staining, while 6/8 were negative. In conclusion, the results support the use of GPNMB to distinguish RCCFMS with TSC1/2/MTOR alterations from others with similar morphology.
Collapse
Affiliation(s)
- Huili Li
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | - Pedram Argani
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | | | - Tamara L. Lotan
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| | - Maria J. Merino
- Translational Surgical Pathology, Laboratory of Pathology, National Institutes of health, Bethesda, MD 20892
| | - Victor E. Reuter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10021
| | - Andres Matoso
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Urology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
- Department of Oncology, The Johns Hopkins Medical Institutions, Baltimore, MD, 21231
| |
Collapse
|
23
|
Kapur P, Brugarolas J, Trpkov K. Recent Advances in Renal Tumors with TSC/mTOR Pathway Abnormalities in Patients with Tuberous Sclerosis Complex and in the Sporadic Setting. Cancers (Basel) 2023; 15:4043. [PMID: 37627070 PMCID: PMC10452688 DOI: 10.3390/cancers15164043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
A spectrum of renal tumors associated with frequent TSC/mTOR (tuberous sclerosis complex/mechanistic target of rapamycin) pathway gene alterations (in both the germline and sporadic settings) have recently been described. These include renal cell carcinoma with fibromyomatous stroma (RCC FMS), eosinophilic solid and cystic renal cell carcinoma (ESC RCC), eosinophilic vacuolated tumor (EVT), and low-grade oncocytic tumor (LOT). Most of these entities have characteristic morphologic and immunohistochemical features that enable their recognition without the need for molecular studies. In this report, we summarize recent advances and discuss their evolving complexity.
Collapse
Affiliation(s)
- Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James Brugarolas
- Kidney Cancer Program at Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hematology-Oncology Division of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB T2L 2K5, Canada
- Alberta Precision Labs, Rockyview General Hospital, 7007 14 St., Calgary, AB T2V 1P9, Canada
| |
Collapse
|
24
|
Batavia AA, Rutishauser D, Sobottka B, Schraml P, Beerenwinkel N, Moch H. Biallelic ELOC-Inactivated Renal Cell Carcinoma: Molecular Features Supporting Classification as a Distinct Entity. Mod Pathol 2023; 36:100194. [PMID: 37088333 DOI: 10.1016/j.modpat.2023.100194] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/28/2023] [Accepted: 04/16/2023] [Indexed: 04/25/2023]
Abstract
Approximately 70% of clear cell renal cell carcinoma (ccRCC) is characterized by the biallelic inactivation of von Hippel-Lindau (VHL) on chromosome 3p. ELOC-mutated (Elongin C-mutated) renal cell carcinoma containing biallelic ELOC inactivations with chromosome 8q deletions is considered a novel subtype of renal cancer possessing a morphologic overlap with ccRCC, renal cell carcinoma (RCC) with fibromyomatous stroma exhibiting Tuberous Sclerosis Complex (TSC)/mammalian Target of Rapamycin (mTOR) mutations, and clear cell papillary tumor. However, the frequency and consequences of ELOC alterations in wild-type VHL and mutated VHL RCC are unclear. In this study, we characterize 123 renal tumors with clear cell morphology and known VHL mutation status to assess the morphologic and molecular consequences of ELOC inactivation. Using OncoScan and whole-exome sequencing, we identify 18 ELOC-deleted RCCs, 3 of which contain ELOC mutations resulting in the biallelic inactivation of ELOC. Biallelic ELOC and biallelic VHL aberrations were mutually exclusive; however, 2 ELOC-mutated RCCs showed monoallelic VHL alterations. Furthermore, no mutations in TSC1, TSC2, or mTOR were identified in ELOC-mutated RCC with biallelic ELOC inactivation. Using High Ambiguity Driven biomolecular DOCKing, we report a novel ELOC variant containing a duplication event disrupting ELOC-VHL interaction alongside the frequently seen Y79C alteration. Using hyper reaction monitoring mass spectrometry, we show RCCs with biallelic ELOC alterations have significantly reduced ELOC expression but similar carbonic anhydrase 9 and vascular endothelial growth factor A expression compared with classical ccRCC with biallelic VHL inactivation. The absence of biallelic VHL and TSC1, TSC2, or mTOR inactivation in RCC with biallelic ELOC inactivation (ELOC mutation in combination with ELOC deletions on chromosome 8q) supports the notion of a novel, molecularly defined tumor entity.
Collapse
Affiliation(s)
- Aashil A Batavia
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dorothea Rutishauser
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bettina Sobottka
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter Schraml
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Niko Beerenwinkel
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland; Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Siadat F, Mansoor M, Hes O, Trpkov K. Kidney Tumors: New and Emerging Kidney Tumor Entities. Clin Lab Med 2023; 43:275-298. [PMID: 37169446 DOI: 10.1016/j.cll.2023.03.003] [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: 05/13/2023]
Abstract
This review summarizes current knowledge on several novel and emerging renal entities, including eosinophilic solid and cystic renal cell carcinoma (RCC), RCC with fibromyomatous stroma, anaplastic lymphoma kinase-rearranged RCC, low-grade oncocytic renal tumor, eosinophilic vacuolated tumor, thyroidlike follicular RCC, and biphasic hyalinizing psammomatous RCC. Their clinical features, gross and microscopic morphology, immunohistochemistry, and molecular and genetic features are described. The diagnosis of most of them rests on recognizing their morphologic features using immunohistochemistry. Accurate diagnosis of these entitles will further reduce the category of "unclassifiable renal carcinomas/tumors" and will lead to better clinical management and improved patient prognostication.
Collapse
Affiliation(s)
- Farshid Siadat
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada. https://twitter.com/FSiadat
| | - Mehdi Mansoor
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada
| | - Ondrej Hes
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, University Hospital Plzen, Alej Svobody 80, 304 60 Pilsen, Czech Republic
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada.
| |
Collapse
|
26
|
Hu X, Tan C, Zhu G. Clinical Characteristics of Molecularly Defined Renal Cell Carcinomas. Curr Issues Mol Biol 2023; 45:4763-4777. [PMID: 37367052 DOI: 10.3390/cimb45060303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Kidney tumors comprise a broad spectrum of different histopathological entities, with more than 0.4 million newly diagnosed cases each year, mostly in middle-aged and older men. Based on the description of the 2022 World Health Organization (WHO) classification of renal cell carcinoma (RCC), some new categories of tumor types have been added according to their specific molecular typing. However, studies on these types of RCC are still superficial, many types of these RCC currently lack accurate diagnostic standards in the clinic, and treatment protocols are largely consistent with the treatment guidelines for clear cell RCC (ccRCC), which might result in worse treatment outcomes for patients with these types of molecularly defined RCC. In this article, we conduct a narrative review of the literature published in the last 15 years on molecularly defined RCC. The purpose of this review is to summarize the clinical features and the current status of research on the detection and treatment of molecularly defined RCC.
Collapse
Affiliation(s)
- Xinfeng Hu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Congzhu Tan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guodong Zhu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
27
|
Kojima F, Musangile FY, Matsuzaki I, Yorita K, Kuroda N, Nagashima Y, Murata SI. Current Knowledge and Prospects for Renal Hemangioblastoma and Renal Cell Carcinoma with Hemangioblastoma-like Features. Biomedicines 2023; 11:biomedicines11051467. [PMID: 37239138 DOI: 10.3390/biomedicines11051467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
Tumors exhibiting histopathological findings similar to those of hemangioblastoma of the central nervous system (CNS-HB) rarely develop in the kidneys. Currently, renal hemangioblastoma (RHB) is considered analogous to CNS-HB; however, they differ in gross appearance, as well as immunohistochemical and molecular findings. In contrast, some renal cell carcinomas reportedly comprise distinct, clear cell renal cell carcinoma (CCRCC)- and hemangioblastoma (HB)-like areas. Initially, renal cell carcinomas with HB-like features (RCC-HBs) were considered a morphological variant of CCRCC owing to their diverse histological findings. However, the immunohistochemical and molecular findings of RCC-HBs suggest that RCC-HB is distinct from CCRCC. Additionally, one of the RCC-HBs had a focal leiomyomatous stroma and TSC2 variant, suggesting that RCC-HB and RCC with fibromyomatous stroma (RCC-FMS) might belong to the same disease entity. Therefore, we comprehensively reviewed the clinical, pathological, and molecular features of RHB, RCC-HB, and the related tumors and discussed the similarities, differences, and relationships between them. We believe that our review would serve as a foundation for further investigation on elucidating the relationship between CNS-HB, RHB, RCC-HB, and RCC-FMS.
Collapse
Affiliation(s)
- Fumiyoshi Kojima
- Department of Human Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Fidele Y Musangile
- Department of Human Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Ibu Matsuzaki
- Department of Human Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Kenji Yorita
- Department of Diagnostic Pathology, Japanese Red Cross Kochi Hospital, 1-4-63-11 Hataminami-cho, Kochi-shi, Kochi 780-8562, Japan
| | - Naoto Kuroda
- Department of Internal Medicine, Kinrou Hospital, 3-2-28 Azounokitamachi, Kochi 781-0011, Japan
| | - Yoji Nagashima
- Department of Surgical Pathology, Tokyo Women's Medical University Hospital, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Shin-Ichi Murata
- Department of Human Pathology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| |
Collapse
|
28
|
Alaghehbandan R, Campbell SC, McKenney JK. Evolution in the Pathologic Classification of Renal Neoplasia. Urol Clin North Am 2023; 50:181-189. [PMID: 36948665 DOI: 10.1016/j.ucl.2023.01.001] [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: 02/22/2023]
Abstract
The pathologic classification of renal tumors is a dynamic and complex process, which has evolved to a "histomolecular" driven system. Despite advances in molecular characterization, most renal tumors can be diagnosed by morphology with or without using a limited set of immunohistochemical stains. If access to molecular resources and specific immunohistochemical markers is limited, pathologists may face difficulties in following an optimal algorithm to classify renal tumors. In this article, we detail the historical evolution of renal tumor classification, including a synopsis of major changes introduced by the current fifth edition World Health Organization 2022 classification of renal epithelial tumors.
Collapse
Affiliation(s)
- Reza Alaghehbandan
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 2119 E. 96th Street, L25, Cleveland, OH 44106, USA
| | - Steven C Campbell
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Q10-120, Glickman Tower, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Jesse K McKenney
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 2119 E. 96th Street, L25, Cleveland, OH 44106, USA.
| |
Collapse
|
29
|
Mimma R, Anna C, Matteo B, Gaetano P, Carlo G, Guido M, Camillo P. Clinico-pathological implications of the 2022 WHO Renal Cell Carcinoma classification. Cancer Treat Rev 2023; 116:102558. [PMID: 37060647 DOI: 10.1016/j.ctrv.2023.102558] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/03/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
The new WHO classification of urogenital tumours published in 2022, contains significant revisions upon the previous 2016 version regarding Renal Cell Carcinoma (RCC). While the most common histotype remains almost untouched, some of the main novelties concerns papillary RCC and oncocytic neoplasms. The main change is the introduction of a new category of molecularly-defined RCC, which includes TFE3-rearranged RCC, TFEB-rearranged, and TFEB-amplified RCC, FH-deficient RCC, SDH-deficient RCC, ALK-rearranged RCC, ELOC (formerly TCEB1)-mutated RCC, SMARCB1 (INI1)-deficient RCC. In this paper we analyze the current knowledge on emerging entities and molecularly-defined RCC to assess whether the current pathological classification offers the oncologist the possibility of selecting more specific and personalized treatments, from both those currently available, as well as those that will soon be available.
Collapse
Affiliation(s)
- Rizzo Mimma
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy.
| | - Caliò Anna
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Brunelli Matteo
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy
| | - Pezzicoli Gaetano
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari "A. Moro", Bari, Italy
| | - Ganini Carlo
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari "A. Moro", Bari, Italy
| | - Martignoni Guido
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Italy; Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Verona, Italy
| | - Porta Camillo
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy; Chair of Oncology, Interdisciplinary Department of Medicine, University of Bari "A. Moro", Bari, Italy
| |
Collapse
|
30
|
Gupta S, Stanton ML, Reynolds JP, Whaley RD, Herrera-Hernandez L, Jimenez RE, Cheville JC. Reprint of: lessons from histopathologic examination of nephrectomy specimens in patients with tuberous sclerosis complex: cysts, angiomyolipomas & renal cell carcinoma. Hum Pathol 2023; 133:136-152. [PMID: 36894367 DOI: 10.1016/j.humpath.2023.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/08/2022] [Indexed: 03/09/2023]
Abstract
Renal manifestations in patients with tuberous sclerosis complex (TSC) include cysts, angiomyolipoma, and renal cell carcinoma. Unlike many hereditary predisposition syndromes, the spectrum of renal tumors in TSC patients (including both angiomyolipoma and renal cell carcinoma) is broad, with significant morphologic heterogeneity. An improved understanding of histopathologic findings in TSC patients and associated clinicopathologic correlates has significant implications not just in establishing a diagnosis of TSC, but also in the recognition of sporadic tumors occurring secondary to somatic alterations of TSC1/TSC2/MTOR pathway genes and accurate prognostication. In this review, we have discussed issues relevant to clinical management based on histopathologic findings in nephrectomy specimens from patients with TSC. This includes discussions related to screening for TSC, diagnosis of PKD1/TSC2 contiguous gene deletion syndrome, the morphologic spectrum of angiomyolipoma and renal epithelium-derived neoplasia, including the risk of disease progression.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Melissa L Stanton
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, 85054, USA.
| | - Jordan P Reynolds
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Rumeal D Whaley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | | | - Rafael E Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
31
|
Zhu S, Chen J, Zeng H. Our Current Understanding of the Heterogeneity in Prostate Cancer and Renal Cell Carcinoma. J Clin Med 2023; 12:jcm12041526. [PMID: 36836061 PMCID: PMC9962000 DOI: 10.3390/jcm12041526] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Urological cancer is a collective term for cancers of the bladder, kidney, ureter, penis, prostate, and testicles. Last year, more than 444,000 people were diagnosed with urinary cancers in the United States. In this review, we talk about the complexity of prostate and kidney cancer.
Collapse
|
32
|
da Paz AR, de Souza MF, Santana CMDM, Athanazio DA. Clear Cell Papillary Renal Cell Tumors: A Study of 42 Tumors with Emphasis on the Fibrous Capsule, Cystic Component, and GATA3 Immunohistochemistry. Int J Surg Pathol 2023; 31:38-45. [PMID: 35503256 DOI: 10.1177/10668969221091583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Clear cell papillary renal cell tumor is a common and sometimes underdiagnosed renal cell neoplasm. Its proper recognition is important because its diagnosis implies a remarkably high probability of indolent behavior. This study aimed to evaluate the frequency of a fibrous capsule, a cystic component, and a GATA3 expression in clear cell papillary renal cell tumors. We assessed 419 renal cell neoplasms from three institutions located in northeastern Brazil and identified 42 clear cell papillary renal cell tumors (from 39 patients), which were the fourth most common renal cell neoplasm. These tumors commonly exhibited fibrous capsules (all showed complete or partial capsules) and cystic component (93%). Eighteen out of 42 tumors (43%) showed some expression of GATA3, and weak and focal staining was common among the positive tumors. Clear cell papillary renal cell tumor must always be included in the differential diagnosis of predominantly cystic renal cell neoplasms. As GATA3 is inconsistently expressed in clear cell papillary renal cell tumors, it is not useful in this diagnosis.
Collapse
Affiliation(s)
- Alexandre Rolim da Paz
- Hospital Napoleão Laureano, João Pessoa, Brazil.,Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Maiara Ferreira de Souza
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Imagepat, Laboratory of Pathology, Brazil
| | | | - Daniel Abensur Athanazio
- Faculdade de Medicina, Universidade Federal da Bahia, Salvador, Brazil.,Imagepat, Laboratory of Pathology, Brazil.,Hospital Universitário Professor Edgard Santos / Federal University of Bahia, Brazil
| |
Collapse
|
33
|
Tjota MY, Sharma A, Wanjari P, Fitzpatrick C, Segal J, Antic T. TSC/MTOR mutated renal cell carcinoma with leiomyomatous stroma is a distinct entity: a comprehensive study of 12 cases. Hum Pathol 2022; 134:124-133. [PMID: 36592877 DOI: 10.1016/j.humpath.2022.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/21/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023]
Abstract
The 2016 World Health Organization (WHO) Classification of Tumors of the Urinary System includes renal cell carcinoma (RCC) with leiomyomatous stroma (RCC-LS) as a provisional category. Recent studies have shown that this category includes at least 4 subtypes: clear cell (CCRCC), clear cell papillary renal cell tumor (CCPRCT), ELOC (TCEB1) mutated, and a subtype of RCC with TSC/MTOR mutations. The most recent 2022 World Health Organization (WHO) Classification of Tumors of the Urinary System includes ELOC mutated RCC-LS as a distinct entity but does not address any other renal tumors with smooth muscle stroma. We reviewed >500 cases of RCC with clear cell phenotype and identified 12 cases that exhibited prominent smooth muscle stroma, of which 4 of the cases had been previously reported. Review of the H&E revealed that all of the tumors were circumscribed with nested, solid, tubular, and tubulopapillary architecture. The epithelium was intimately embedded in the rich smooth muscle stroma. WHO/ISUP grade corresponded to grade 3 and 4. Nuclei were randomly distributed and the cytoplasm had predominantly clear and occasionally flocculent appearance. Immunohistochemically, all the cases showed membranous CAIX staining, although the pattern was combined cup and box-shaped. CK7 was positive in all cases ranging from 25% to 100% of cells. Membranous and apical staining of CD10 was present in all cases. Next generation sequencing (NGS) of these cases identified mutations in TSC1 (n = 4), TSC2 (n = 3), and MTOR (n = 4) with one case exhibiting loss of TSC1. This descriptive study, although small, demonstrates the difficulty in applying the current WHO provisional criteria at a single institution. Given the heterogeneity seen with these cases, we suggest following up an immunohistochemical panel of CAIX, CK7, and CD10 with molecular diagnostic studies to assist in the diagnosis of TSC/MTOR associated RCC-LS, which we believe is a distinct entity.
Collapse
Affiliation(s)
- Melissa Y Tjota
- Department of Pathology, The University of Chicago, IL, 60605, USA
| | - Aarti Sharma
- Department of Pathology, The University of Chicago, IL, 60605, USA
| | - Pankhuri Wanjari
- Department of Pathology, The University of Chicago, IL, 60605, USA
| | | | - Jeremy Segal
- Department of Pathology, The University of Chicago, IL, 60605, USA
| | - Tatjana Antic
- Department of Pathology, The University of Chicago, IL, 60605, USA.
| |
Collapse
|
34
|
Are Renal Cell Carcinoma with Fibromyomatous Stroma (RCC-FMS) and Thyroid-like Follicular Carcinoma of the Kidney (TLFCK) Really Independent Variants? Diagnostics (Basel) 2022; 13:diagnostics13010086. [PMID: 36611378 PMCID: PMC9818596 DOI: 10.3390/diagnostics13010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/16/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Renal cell carcinoma with fibromyomatous stroma (RCC-FMS) is a recent provisional entity already recognised in the 2016 WHO Classification of Cancer of the Urinary Tract and Male Genital Organs 4th Edition as renal cell carcinoma with (angio)leiomyomatous stroma, histologically defined as a tumour characterised by clear cells intertwined in a conspicuous vascular stroma. In the casuistry taken into consideration, another proposed variant, thyroid-like follicular carcinoma of the kidney (TLFCK), endowed with a morphology mimicking thyroid parenchyma, was examined. The aim of this work was to parse the theoretical system, experimental data and diagnostic impact of these new entities proposed in the field of renal neoplasms. MATERIALS AND METHODS An analysis of 120 cases of kidney tumours from the Department of Surgical, Medical, Molecular and Critical Area at the University of Pisa was run. Subsequently, all samples were reassessed by two pathologists with expertise in uropathology, whose revaluation provided a histomorphological study combined with subsequent and coherent immunohistochemical analyses of CK7, CD10, CAIX, CK34betaE12, CD117, vimentin, TTF-1 and thyroglobulin. These analyses were performed using the Ventana Benchmark Automated Staining System (Ventana Medical Systems, Tucson, AZ, USA) and Ventana reagents. RESULTS On the one hand, the data, thus brought to light, did not show an immunohistochemical profile consistent with that proposed for RCC-FMS. However, it should be emphasised that the morphological background also unearthed a poor specificity for RCC-FMS. This was specifically due to a stromal component which was, in any case, evident, although characterised by a wide range of presentation, in clear cell renal cell carcinoma (ccRCC). This latter is, indeed, the reference background for this theorised variant. On the other hand, a thyroid-like pattern was highlighted in 11 cases, more specifically in 10 ccRCCs and in one oncocytoma, presenting itself as a type of neoplastic appearance rather than as the peculiar morphological pattern of a standalone cancer. CONCLUSIONS In the light of these results, RCC-FMS and TLFCK appear to be more appropriately variants of already categorised neoplastic entities rather than new independent neoplasias.
Collapse
|
35
|
Demko N, Glennon KI, Arseneault M, Lach K, Nishimura T, Tanguay S, Riazalhosseini Y, Brimo F. A Sarcomatoid Renal Cell Carcinoma with Clear Cell Papillary-Like Primary Tumor and Lymph Node Metastasis: A Diagnostic Conundrum. Int J Surg Pathol 2022:10668969221145011. [PMID: 36562104 DOI: 10.1177/10668969221145011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Clear cell papillary renal cell tumor (CCPRCT) is a distinct clinical entity with characteristic pathological features and non-aggressive clinical behavior. Diagnostically challenging cases present when there are immunomorphological findings of CCPRCT associated with heterogeneous morphologies, aggressive histological features, and advanced pathological stages-so-called CCPRCT-like tumors. In this report, we describe a heterogeneous, multifocal renal tumor with immunomorphological characteristics of CCPRCT but with associated aggressive features such as sarcomatoid and necrotic areas, perirenal and sinus fat involvement, and most notably, lymph node metastasis composed entirely of classic CCPRCT morphology and immunophenotype. Immunohistochemical and fluorescence in situ hybridization studies did not support a translocation renal cell carcinoma. Molecular analyses did not identify common mutations or chromosomal abnormalities seen in clear cell renal cell carcinoma or ELOC-mutated renal cell carcinoma. This case highlights that rare renal cell tumors remain difficult to classify and the distinction between CCPRCT and CCPRCT-like tumors remains to be better defined.
Collapse
Affiliation(s)
- Nadine Demko
- Department of Anatomical Pathology, 5620McGill University, Montréal, Québec, Canada
| | - Kate I Glennon
- Department of Human Genetics, 5620McGill University, Montréal, Québec, Canada.,McGill Genome Centre, 5620McGill University, Montréal, Québec, Canada
| | - Madeleine Arseneault
- Department of Human Genetics, 5620McGill University, Montréal, Québec, Canada.,McGill Genome Centre, 5620McGill University, Montréal, Québec, Canada
| | - Katherine Lach
- Department of Anatomical Pathology, 5620McGill University, Montréal, Québec, Canada
| | - Tamiko Nishimura
- Department of Human Genetics, 5620McGill University, Montréal, Québec, Canada.,McGill Genome Centre, 5620McGill University, Montréal, Québec, Canada
| | - Simon Tanguay
- Division of Urology, 5620McGill University, Montréal, Québec, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, 5620McGill University, Montréal, Québec, Canada.,McGill Genome Centre, 5620McGill University, Montréal, Québec, Canada
| | - Fadi Brimo
- Department of Anatomical Pathology, 5620McGill University, Montréal, Québec, Canada
| |
Collapse
|
36
|
Kong J, Tao J, Wang Q, Zhang Q, Yin L. Rare renal cell carcinoma with haemangioblastoma-like features and leiomyomatous stroma: report of a unique case with TSC2 and SETD2 variations. World J Surg Oncol 2022; 20:395. [PMID: 36510186 PMCID: PMC9746168 DOI: 10.1186/s12957-022-02844-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/20/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Renal cell carcinoma (RCC) with haemangioblastoma-like characteristics is a type of RCC reported in recent years. RCC with (angio) leiomyomatous stroma (RCCLMS) was included as a provisional entity of the 2016 World Health Organization (WHO) classification. RCC with haemangioblastoma-like characteristics and leiomyomatous stroma is extremely rare. This is the first report of a rare tumour harbouring TSC2 and SETD2 variations. CASE PRESENTATION The patient was a 38-year-old woman who presented with discomfort in the area of her right kidney. Ultrasound and enhanced CT showed a right renal mass, and clear cell renal cell carcinoma (CCRCC) was suspected; hence, robot-assisted laparoscopic nephron-sparing partial nephrectomy was performed. Gross examination revealed a well-circumscribed tumour measuring 2.0 cm × 1 cm × 0.7 cm under the renal capsule adjacent to the stripping edge that was greyish yellow and greyish red in colour. Histologic examination showed that the tumour consisted of three different structures: a CCRCC-like region, a haemangioblastoma-like region, and a focal leiomyomatous stroma component. Based on immunohistochemistry, the CCRCC-like region was diffusely strongly positive for AE1/AE3, vimentin, CAIX, PAX8, PAX2, CK7, and CAM5.2, partly positive for HNF1α, and negative for CD10, α-inhibin, NSE, S-100, CD34, and TFE3. The haemangioblastoma-like area was diffusely positive for vimentin, CAIX; partly positive for PAX8, PAX2, α-inhibin, and S-100; mostly positive for NSE; and slightly positive for HNF1α; the CD34 staining highlighted the complex capillary network. The Ki67 index was approximately 1-2% in the two above areas, and the leiomyomatous stroma was strongly positive for SMA. The whole-exon sequencing (WES) showed TSC2 and SETD2 variations. There was no progression after 18 months of follow-up. CONCLUSION We report for the first time a unique case of RCC with haemangioblastoma-like features and leiomyomatous stroma accompanied by rare molecular abnormalities. Whether this is a new tumour entity or a variant of clear cell carcinoma remains to be determined. The biological behaviour and clinical characteristics need to be further examined.
Collapse
Affiliation(s)
- Jixia Kong
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Juan Tao
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Qimin Wang
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| | - Qingfu Zhang
- grid.412636.40000 0004 1757 9485Department of Pathology, the First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning China
| | - Liying Yin
- grid.452828.10000 0004 7649 7439Department of Pathology, the Second Affiliated Hospital of Dalian Medical University, Dalian, 116027 Liaoning China
| |
Collapse
|
37
|
Muacevic A, Adler JR, Ghoundale O, Azami MA. Renal Cell Carcinoma With Fibromyomatous Stroma: A New Case. Cureus 2022; 14:e32238. [PMID: 36620787 PMCID: PMC9815789 DOI: 10.7759/cureus.32238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
In the 2016 World Health Organization (WHO) classification of renal tumors, renal cell carcinoma with fibromyomatous stroma (RCC FMS) (formerly RCC with leiomyomatous or smooth muscle stroma) was classified as an emerging or provisional entity of renal cell carcinoma (RCC). We report a rare case of RCC FMS in a 62-year-old male patient with hypertension, type II diabetes mellitus, and early chronic kidney disease. He was referred to the Department of Urology for an incidental finding of a 2-cm-long left renal nodule on a routine abdominal ultrasound. A laparoscopic right partial nephrectomy was performed. Histopathology and immunohistochemistry studies confirm the diagnosis of RCC FMS. The purpose of this work is to review and discuss newly acquired data and evidence on the clinical, pathological, immunohistochemical, molecular, and prognostic aspects of this unusual entity in the hopes of assisting pathologists in accurate diagnosis.
Collapse
|
38
|
Siadat F, Mansoor M, Hes O, Trpkov K. Kidney Tumors: New and Emerging Kidney Tumor Entities. Surg Pathol Clin 2022; 15:713-728. [PMID: 36344185 DOI: 10.1016/j.path.2022.07.006] [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] [Indexed: 05/18/2023]
Abstract
This review summarizes current knowledge on several novel and emerging renal entities, including eosinophilic solid and cystic renal cell carcinoma (RCC), RCC with fibromyomatous stroma, anaplastic lymphoma kinase-rearranged RCC, low-grade oncocytic renal tumor, eosinophilic vacuolated tumor, thyroidlike follicular RCC, and biphasic hyalinizing psammomatous RCC. Their clinical features, gross and microscopic morphology, immunohistochemistry, and molecular and genetic features are described. The diagnosis of most of them rests on recognizing their morphologic features using immunohistochemistry. Accurate diagnosis of these entitles will further reduce the category of "unclassifiable renal carcinomas/tumors" and will lead to better clinical management and improved patient prognostication.
Collapse
Affiliation(s)
- Farshid Siadat
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada. https://twitter.com/FSiadat
| | - Mehdi Mansoor
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada
| | - Ondrej Hes
- Department of Pathology, Charles University in Prague, Faculty of Medicine in Plzeň, University Hospital Plzen, Alej Svobody 80, 304 60 Pilsen, Czech Republic
| | - Kiril Trpkov
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Rockyview General Hospital, 7007 14 Street, Calgary, Alberta T2V 1P9, Canada.
| |
Collapse
|
39
|
Akgul M, Williamson SR. How New Developments Impact Diagnosis in Existing Renal Neoplasms. Surg Pathol Clin 2022; 15:695-711. [PMID: 36344184 DOI: 10.1016/j.path.2022.07.005] [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] [Indexed: 06/16/2023]
Abstract
In recent years, several emerging diagnostic entities have been described in renal cell carcinoma (RCC). However, our understanding of well-known and established entities has also grown. Clear cell papillary RCC is now relabeled as a tumor rather than carcinoma in view of its nonaggressive behavior. Renal tumors with a predominantly infiltrative pattern are very important for recognition, as most of these have aggressive behavior, including fumarate hydratase-deficient RCC, SMARCB1-deficient medullary carcinoma, collecting duct carcinoma, urothelial carcinoma, and metastases from other cancers.
Collapse
Affiliation(s)
- Mahmut Akgul
- Department of Pathology and Laboratory Medicine, Albany Medical Center, 47 New Scotland Ave, Room F110S, MC81 Albany, NY 12208, USA
| | - Sean R Williamson
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Mail Code L25 Cleveland, OH 44195, USA.
| |
Collapse
|
40
|
Gupta S, Sukov WR, Vanderbilt CM, Shen W, Herrera-Hernandez L, Lohse CM, Thompson RH, Boorjian SA, Leibovich BC, Jimenez RE, Cheville JC. A contemporary guide to chromosomal copy number profiling in the diagnosis of renal cell carcinoma. Urol Oncol 2022; 40:512-524. [PMID: 34092479 DOI: 10.1016/j.urolonc.2021.04.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/06/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
The routine clinical implementation of molecular methods other than fluorescence in situ hybridization in the evaluation of renal neoplasia is currently limited, as the current standard of care primarily involves a combination of morphologic and immunophenotypic analysis of such tumors. Amongst various molecular techniques, global copy number profiling using single nucleotide polymorphism-based microarrays, colloquially referred to as SNP-arrays, is being increasingly utilized to profile renal tumors, as several subtypes have characteristic recurrent patterns of copy number alterations. Recurrent copy number alterations in common tumor types include loss of chromosome 3p in clear cell renal cell carcinoma (RCC), gain of chromosomes 7 and 17 in papillary RCC and multiple losses in chromosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe RCC. Such assays are being increasingly utilized in the clinical setting. Herein, we discuss some common clinical applications of such testing that includes high yield diagnostic and prognostic applications. Diagnostic utility includes evaluation of tumor types that are primarily defined by underlying copy number alterations, establishing the underlying subtype in high grade dedifferentiated (unclassified) renal tumors, as well as assessment of loss of heterozygosity, which is an important component in the workup for germline alterations in tumor suppressor genes. Universal adoption of these techniques across clinical laboratories will likely be significantly affected by variables such as cost, reimbursement, and turnaround time.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN.
| | - William R Sukov
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Wei Shen
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Christine M Lohse
- Department of Quantitative Health Sciences, 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
|
41
|
Argani P, Rose G, Matoso A, Gagan J, Palsgrove DN. Biphasic Hyalinizing Psammomatous Renal Cell Carcinoma (BHP RCC) in a Child With Neurofibromatosis Type 2 Syndrome. Am J Surg Pathol 2022; 46:1595-1598. [PMID: 35941722 PMCID: PMC11841680 DOI: 10.1097/pas.0000000000001942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Pedram Argani
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gary Rose
- Department of Pathology, Saint Joseph's Hospital, BayCare Laboratories, Tampa FL
| | - Andres Matoso
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jeffrey Gagan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Doreen N Palsgrove
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
42
|
Gupta S, Stanton ML, Reynolds JP, Whaley RD, Herrera-Hernandez L, Jimenez RE, Cheville JC. Lessons from histopathologic examination of nephrectomy specimens in patients with tuberous sclerosis complex: cysts, angiomyolipomas, and renal cell carcinoma. Hum Pathol 2022; 129:123-139. [PMID: 36115585 DOI: 10.1016/j.humpath.2022.09.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/08/2022] [Indexed: 02/07/2023]
Abstract
Renal manifestations in patients with tuberous sclerosis complex (TSC) include cysts, angiomyolipoma, and renal cell carcinoma. Unlike many hereditary predisposition syndromes, the spectrum of renal tumors in TSC patients (including both angiomyolipoma and renal cell carcinoma) is broad, with significant morphologic heterogeneity. An improved understanding of histopathologic findings in TSC patients and associated clinicopathologic correlates has significant implications not just in establishing a diagnosis of TSC, but also in the recognition of sporadic tumors occurring secondary to somatic alterations of TSC1/TSC2/MTOR pathway genes and accurate prognostication. In this review, we have discussed issues relevant to clinical management based on histopathologic findings in nephrectomy specimens from patients with TSC. This includes discussions related to screening for TSC, diagnosis of PKD1/TSC2 contiguous gene deletion syndrome, the morphologic spectrum of angiomyolipoma and renal epithelium-derived neoplasia, including the risk of disease progression.
Collapse
Affiliation(s)
- Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Melissa L Stanton
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ, 85054, USA.
| | - Jordan P Reynolds
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Rumeal D Whaley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | | | - Rafael E Jimenez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - John C Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
43
|
Moch H, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, Menon S, Raspollini MR, Rubin MA, Srigley JR, Hoon Tan P, Tickoo SK, Tsuzuki T, Turajlic S, Cree I, Netto GJ. The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours. Eur Urol 2022; 82:458-468. [PMID: 35853783 DOI: 10.1016/j.eururo.2022.06.016] [Citation(s) in RCA: 314] [Impact Index Per Article: 104.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 02/07/2023]
Abstract
The fifth edition of the World Health Organization (WHO) classification of urogenital tumours (WHO "Blue Book"), published in 2022, contains significant revisions. This review summarises the most relevant changes for renal, penile, and testicular tumours. In keeping with other volumes in the fifth edition series, the WHO classification of urogenital tumours follows a hierarchical classification and lists tumours by site, category, family, and type. The section "essential and desirable diagnostic criteria" included in the WHO fifth edition represents morphologic diagnostic criteria, combined with immunohistochemistry and relevant molecular tests. The global introduction of massive parallel sequencing will result in a diagnostic shift from morphology to molecular analyses. Therefore, a molecular-driven renal tumour classification has been introduced, taking recent discoveries in renal tumour genomics into account. Such novel molecularly defined epithelial renal tumours include SMARCB1-deficient medullary renal cell carcinoma (RCC), TFEB-altered RCC, Alk-rearranged RCC, and ELOC-mutated RCC. Eosinophilic solid and cystic RCC is a novel morphologically defined RCC entity. The diverse morphologic patterns of penile squamous cell carcinomas are grouped as human papillomavirus (HPV) associated and HPV independent, and there is an attempt to simplify the morphologic classification. A new chapter with tumours of the scrotum has been introduced. The main nomenclature of testicular tumours is retained, including the use of the term "germ cell neoplasia in situ" (GCNIS) for the preneoplastic lesion of most germ cell tumours and division from those not derived from GCNIS. Nomenclature changes include replacement of the term "primitive neuroectodermal tumour" by "embryonic neuroectodermal tumour" to separate these tumours clearly from Ewing sarcoma. The term "carcinoid" has been changed to "neuroendocrine tumour", with most examples in the testis now classified as "prepubertal type testicular neuroendocrine tumour".
Collapse
Affiliation(s)
- Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zuerich and University of Zuerich, Zuerich, Switzerland.
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Urology, USC Keck School of Medicine, Los Angeles, CA, USA
| | - Daniel M Berney
- Barts Cancer Institute, Queen Mary University of London, London, UK; Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Eva M Compérat
- Department of Pathology, Medical University of Vienna, General Hospital of Vienna, Vienna, Austria
| | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney, Australia; NSW Health Pathology, Department of Anatomical Pathology and Pathology Group Kolling Institute of Medical Research Royal North Shore Hospital St Leonards, Sydney, Australia
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Santosh Menon
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Maria R Raspollini
- Histopathology and Molecular Diagnostics, University Hospital Careggi, Florence, Italy
| | - Mark A Rubin
- Department for BioMedical Research (DBMR), Bern Center for Precision Medicine (BCPM), University of Bern and Inselspital, Bern, Switzerland
| | - John R Srigley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| | - Satish K Tickoo
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Toyonori Tsuzuki
- Department of Surgical Pathology, Aichi Medical University Hospital, Nagakut, Japan
| | - Samra Turajlic
- The Francis Crick Institute and The Royal Marsden NHS Foundation Trust, London, UK
| | - Ian Cree
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France
| | - George J Netto
- Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
44
|
French AFU Cancer Committee Guidelines - Update 2022-2024: management of kidney cancer. Prog Urol 2022; 32:1195-1274. [DOI: 10.1016/j.purol.2022.07.146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 07/14/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
|
45
|
Do we need an updated classification of oncocytic renal tumors? : Emergence of low-grade oncocytic tumor (LOT) and eosinophilic vacuolated tumor (EVT) as novel renal entities. Mod Pathol 2022; 35:1140-1150. [PMID: 35273336 DOI: 10.1038/s41379-022-01057-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 02/07/2023]
Abstract
The category of "oncocytic renal tumors'' includes well-recognized entities, such as renal oncocytoma (RO) and eosinophilic variant of chromophobe renal cell carcinoma (eo-ChRCC), as well as a group of "gray zone" oncocytic tumors, with overlapping features between RO and eo-ChRCC that create ongoing diagnostic and classification problems. These types of renal tumors were designated in the past as "hybrid oncocytoma-chromophobe tumors". In a recent update, the Genitourinary Pathology Society (GUPS) proposed the term "oncocytic renal neoplasm of low malignant potential, not further classified", for such solitary and sporadic, somewhat heterogeneous, but relatively indolent tumors, with equivocal RO/eo-ChRCC features. GUPS also proposed that the term "hybrid oncocytic tumor" be reserved for tumors found in a hereditary setting, typically arising as bilateral and multifocal ones (as in Birt-Hogg-Dubé syndrome). More recent developments in the "gray zone" of oncocytic renal tumors revealed that potentially distinct entities may have been "hidden" in this group. Recent studies distinguished two new entities: "Eosinophilic Vacuolated Tumor" (EVT) and "Low-grade Oncocytic Tumor" (LOT). The rapidly accumulated evidence on EVT and LOT has validated the initial findings and has expanded the knowledge on these entities. Both are uniformly benign and are typically found in a sporadic setting, but rarely can be found in patients with tuberous sclerosis complex. Both have readily distinguishable morphologic and immunohistochemical features that separate them from similar renal tumors, without a need for detailed molecular studies. These tumors very frequently harbor TSC/MTOR mutations that are however neither specific nor restricted to these two entities. In this review, we outline a proposal for a working framework on how to classify such low-grade oncocytic renal tumors. We believe that such framework will facilitate their handling in practice and will stimulate further discussions and studies to fully elucidate their spectrum.
Collapse
|
46
|
Xanthomatous Giant Cell Renal Cell Carcinoma: Another Morphologic Form of TSC-associated Renal Cell Carcinoma. Am J Surg Pathol 2022; 46:1554-1561. [PMID: 35941720 DOI: 10.1097/pas.0000000000001940] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Over the past decade, several distinct novel renal epithelial neoplasms driven by underlying tuberous sclerosis comples (TSC)/mammalian target of rapamycin (MTOR) pathway mutations have been described. We report herein two distinctive TSC2-mutated renal cell carcinomas which do not fit any previously described entity. The two renal carcinomas occurred in young patients (ages 10 and 31 y), and were characterized by highly permeative growth within the kidney with metastases to perirenal lymph nodes. The neoplastic cells were predominantly large, multinucleated giant cells having variably eosinophilic to xanthomatous cytoplasm with basophilic stippling and frequent vacuolization. While the discohesive nature of the neoplastic cells, xanthomatous cytoplasm, immunoreactivity for histiocytic markers and minimal immunoreactivity for conventional epithelial markers raised the possibility of a histiocytic neoplasm, multifocal immunoreactivity for cytokeratin 20 helped establish their epithelial nature. Despite the aggressive growth pattern of these neoplasms and lymph node metastases, mitotic figures were rare and Ki-67 indices were low (<1%). One patient with follow-up shows no evidence of disease seven years after nephrectomy with no adjuvant therapy. Next-generation sequencing demonstrated TSC2 mutations in each case. By immunohistochemistry, downstream markers of mTOR pathway activation S6K1, 4EBP1, and glycoprotein nonmetastatic melanoma protein B were all highly expressed in these neoplasms, suggesting mTOR pathway activation as the neoplastic driver. While the cytokeratin 20 immunoreactivity and focal basophilic cytoplasmic stippling suggest a relationship to eosinophilic solid and cystic renal cell carcinoma, and cytoplasmic vacuolization suggests a relationship to eosinophilic vacuolated tumor, these neoplasms appear to be distinctive given their permeative growth patterns and predominant xanthomatous giant cell morphology. Addition of cytokeratin 20 to a panel of epithelial markers helps avoid misdiagnosis in such cases.
Collapse
|
47
|
Wang Y, Zhao P, Wang L, Wang J, Ji X, Li Y, Shi H, Li Y, Zhang W, Jiang Y. Analysis of clinicopathological and molecular features of ELOC(TCEB1)-mutant renal cell carcinoma. Pathol Res Pract 2022; 235:153960. [DOI: 10.1016/j.prp.2022.153960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/10/2022] [Accepted: 05/25/2022] [Indexed: 11/28/2022]
|
48
|
Abstract
Renal cell carcinoma (RCC) with fibromyomatous stroma (FMS) was included as an "emerging/provisional" entity in the 2016 World Health Organization (WHO) classification as a "RCC with (angio) leiomyomatous stroma." It has been debated whether RCCFMS represents a separate entity or a group of RCCs with overlapping morphologies. Accordingly, various names have been used to refer to the RCCs that exhibited clear cells and prominent smooth muscle and fibromatous stroma. Recent studies have demonstrated that RCCFMS indeed represents a distinct entity with subtle but distinguishable features that can be separated from other RCCs that exhibit clear cells, as well as tubulopapillary morphology and smooth muscle/fibromatous stroma, such as clear cell RCC and clear cell papillary RCC. Microscopically, the epithelial component forms tumor nodules composed of elongated and frequently branching tubules, lined by clear or mildly eosinophilic cells containing voluminous cytoplasm. Focal papillary morphology is also frequently present. Diffuse CK7 positivity is typical and is required for the diagnosis. Molecular analysis of these tumors demonstrated recurrent mutations involving the TSC/mTOR pathway. A subset of tumors with similar morphology has shown mutations involving ELOC (previously referred to as TCEB1), typically associated with monosomy 8. Finally, in addition to the more common RCCFMS that are sporadic, essentially identical tumors have been found in patients with tuberous sclerosis complex, suggesting the existence of hereditary and sporadic counterparts of this tumor. It is currently debated whether TSC/mTOR and ELOC mutated RCCFMS should be grouped together, based on their shared and overlapping morphology and common CK7 reactivity, despite the differing molecular alterations. This review outlines evidence supporting the recognition of RCCFMS as a novel subtype of RCC with morphologic, immunohistochemical, and molecular characteristics distinct from clear cell RCC and clear cell papillary RCC.
Collapse
Affiliation(s)
- Rajal B Shah
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
49
|
Paner GP, Chumbalkar V, Montironi R, Moch H, Amin MB. Updates in Grading of Renal Cell Carcinomas Beyond Clear Cell Renal Cell Carcinoma and Papillary Renal Cell Carcinoma. Adv Anat Pathol 2022; 29:117-130. [PMID: 35275846 DOI: 10.1097/pap.0000000000000341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The World Health Organization (WHO) recommends grading of clear cell renal cell carcinoma (RCC) and papillary RCC using the WHO/International Society of Urological Pathology (ISUP) grade, which is primarily based on nuclear features. As the spectrum of RCC continues to evolve, with more recently described subtypes in the past decade, literature evidence on grading these subtypes is limited or not available for some tumor types. Herein, we outline a pragmatic approach to the topic of grading RCC, dividing the contemporarily described RCC subtypes into 7 categories based on the potential clinical applicability of grading as a useful prognostic parameter: (1) RCC subtypes that are reasonably validated and recommended for WHO/ISUP grading; (2) RCC subtypes where WHO/ISUP is not applicable; (3) RCC subtypes where WHO/ISUP grading is potentially clinically useful; (4) inherently aggressive RCC subtypes where histologic classification itself confers an aggressive biologic potential; (5) renal epithelial tumors where WHO/ISUP grading provides potentially misleading prognostic implication; (6) renal epithelial neoplasms where low WHO/ISUP grade features are a prerequisite for accurate histologic classification; and (7) renal epithelial neoplasms with no or limited data on grading or incomplete understanding of the biologic potential. Our aim in outlining this approach is 2-fold: (a) identify the gaps in understanding and application of grading in RCC subtypes so that researchers in the field may perform additional studies on the basis of which the important pathologic function of assignment of grade may be recommended to be performed as a meaningful exercise across a wider spectrum of RCC; and (b) to provide guidance in the interim to surgical pathologists in terms of providing clinically useful grading information in RCC based on currently available clinicopathologic information.
Collapse
Affiliation(s)
- Gladell P Paner
- Department of Pathology, University of Chicago
- Department of Surgery, Section of Urology, University of Chicago, Chicago, IL
| | | | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical and Molecular Sciences, Polytechnic University of the Marche Region, Ancona, Italy
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN
- Department of Urology, USC Keck School of Medicine, Los Angeles, CA
| |
Collapse
|
50
|
Lobo J, Rechsteiner M, Helmchen BM, Rupp NJ, Weber A, Moch H. Eosinophilic solid and cystic renal cell carcinoma and renal cell carcinomas with TFEB alterations: a comparative study. Histopathology 2022; 81:32-43. [PMID: 35403742 DOI: 10.1111/his.14663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/23/2022] [Accepted: 04/10/2022] [Indexed: 11/27/2022]
Abstract
AIMS Eosinophilic solid and cystic renal cell carcinoma (ESC RCC) is a recently described renal tumour entity with frequent CK20 positivity, commonly harbouring TSC mutations. In contrast, frequency of CK20 expression and presence of TSC mutations are unclear in TFEB-amplified RCC and TFEB-translocated RCC, which frequently express Melan A. Herein, we provide a comparative analysis of 6 ESC RCC with 4 TFEB-amplified/translocated RCC. METHODS AND RESULTS We assessed the frequency of CK20 and Melan A expression by immunohistochemistry, and of TSC mutations by next generation sequencing. TFEB alterations were confirmed by fluorescence in situ hybridization (FISH). All tumours showed voluminous eosinophilic cells with granular cytoplasm, prominent nucleoli, and most showed admixture of solid and cystic areas. CK20 expression was found in all 6 ESC RCC and in all RCCs with TFEB alterations. Melan A positivity was identified in 5/6 ESC RCC and 4/4 RCC with TFEB alterations. We found TSC mutations in 2 ESC RCCs, including in one case also harbouring a CIC fusion, and identified a TSC mutation in one TFEB-amplified RCC. CONCLUSIONS ESC RCC represents an emerging renal tumour entity with some histological, immunohistochemical and molecular overlap to TFEB-amplified/translocated RCC. FISH for TFEB aids in this differential diagnosis in challenging cases.
Collapse
Affiliation(s)
- João Lobo
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,Cancer Biology and Epigenetics Group, IPO Porto Research Center (GEBC CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto) & Porto Comprehensive Cancer Center (P.CCC), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Markus Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Birgit M Helmchen
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006, Zurich, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, CH 8091, Zurich, Switzerland.,Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006, Zurich, Switzerland
| |
Collapse
|