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; 38:100777. [PMID: 40246078 DOI: 10.1016/j.modpat.2025.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 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 RCC first recognized by the World Health Organization in 2022, molecularly defined by the presence of ELOC mutations and the lack of VHL mutations. Here, we present an institutional series of ELOC-RCC and provide an in-depth genetic comparison to VHL-null clear cell RCC (VHL-ccRCCs). Among 1209 RCCs in our institutional cytogenetics database, we identified 16 candidate cases that were originally classified as 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 6 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, and 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, 0.32; 95% confidence intervals, 0.16-0.61; P = .02) and progression-free survival (hazard ratio, 0.16; 95% confidence intervals, 0.06-0.42; P = 0.04). ELOC-RCCs had significantly fewer somatic copy number alterations and a 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, whereas additional alterations to ELOC or 8q losses occurred as a subclonal event. Although 11 of 13 ELOC-RCCs were confined to the kidney, 2 ELOC-RCCs were high-stage and exhibited a large solid alveolar pattern, tumor necrosis, more somatic copy number alterations, 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, California
| | - Rong Rong Huang
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Brian D Cone
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Sung-Hae L Kang
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Reza Setoodeh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Anthony E Sisk
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Dipti P Sajed
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California
| | - Brian M Shuch
- Department of Urology, University of California Los Angeles, David Geffen School of Medicine, Los Angeles, California.
| | - Adam G Sowalsky
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, Maryland.
| | - Huihui Ye
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
2
|
Albertson D, Barry M, Liu T, Mahlow J, Sirohi D. Genotype Phenotype Correlation of Renal Tumors in the Cancer Genome Atlas Database. Int J Surg Pathol 2025; 33:289-301. [PMID: 39034146 DOI: 10.1177/10668969241260236] [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: 07/23/2024]
Abstract
Morphological features are critical in evaluation of renal tumors and directing molecular workup. The objective of this study was to review histomorphology of renal tumors with molecular alterations of known subtypes. Renal tumors in The Cancer Genome Atlas were reviewed to identify tumors with defining molecular alterations. Single representative digital slides and pathology reports were reviewed and morphologic features recorded. Sixty tumors were identified with molecular alterations in genes characteristic of defined renal cell carcinoma (RCC) subtypes. Findings included the presence of both low- and high-grade histology in TFE3 rearranged RCCs, TFEB amplified RCCs, succinate dehydrogenase (SDH) mutated RCCs and RCCs with mutations in mismatch repair genes. Three ELOC mutated RCCs were identified, one of which demonstrated infiltrative features. Pseudostratification of nuclei in fumarate hydratase mutated RCCs and nuclear grooves in SDH mutated RCCs were intriguing findings not previously reported. Mucinous features were noted in NF2, KRAS, and SDH mutated and ALK rearranged tumors. Significant morphologic overlap was noted across most categories with limited clues for subclassification. Whereas the number of diagnostic entities for kidney tumors continues to increase, many of these have overlapping features, highlighting the significant role molecular characterization currently plays and will continue to play in the future.
Collapse
Affiliation(s)
- Daniel Albertson
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Marc Barry
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Ting Liu
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Jonathan Mahlow
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| | - Deepika Sirohi
- Department of Pathology, University of Utah and ARUP Laboratories, Salt Lake City, UT, USA
| |
Collapse
|
3
|
Nezami BG, MacLennan GT. Clear Cell Renal Cell Carcinoma: A Comprehensive Review of its Histopathology, Genetics, and Differential Diagnosis. Int J Surg Pathol 2025; 33:265-280. [PMID: 39051572 DOI: 10.1177/10668969241256111] [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: 07/27/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the predominant subtype of renal epithelial tumor, accounting for roughly 2% of all malignancies. Clinically, it often presents in the sixth to seventh decade of life, predominantly in men. Pathologically, these tumors exhibit a distinctive golden yellow cut surface, usually arising from the renal cortex. Their microscopic features are characterized by solid and nested architectures of cells with clear or eosinophilic granular cytoplasm and a prominent vascular network. A hallmark genetic feature is the inactivation of the VHL gene situated on chromosome 3p25. The majority of ccRCCs are sporadic (over 95%), typically presenting as a single mass; and a small percentage have a hereditary basis, often associated with VHL disease, characterized by multiple bilateral tumors with an earlier onset. Immunohistochemically, ccRCC tumors express PAX8, CA9 box like pattern, and CD10 but are generally negative for AMACR (35% positive) and KRT7 (15% positive). The prognosis of ccRCC is largely determined by its TNM stage, ISUP/WHO nucleolar grade, and the presence of specific aggressive features. This review article delves into the detailed gross, microscopic, molecular, and clinical features of ccRCC, offering comprehensive insights into its diagnosis, management, and prognosis.
Collapse
Affiliation(s)
- Behtash G Nezami
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Gregory T MacLennan
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhu B, Liu S, David NL, Dion W, Doshi NK, Siegel LB, Amorim T, Andrews RE, Kumar GVN, Li H, Irfan S, Pesaresi T, Sharma AX, Sun M, Fazeli PK, Steinhauser ML. Evidence for ~12-h ultradian gene programs in humans. NPJ BIOLOGICAL TIMING AND SLEEP 2024; 1:4. [PMID: 39148626 PMCID: PMC11325440 DOI: 10.1038/s44323-024-00005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024]
Abstract
Mice and many marine organisms exhibit ~12-h ultradian rhythms, however, direct evidence of ~12-h ultradian rhythms in humans is lacking. Here, we performed prospective, temporal transcriptome profiling of peripheral white blood cells from three healthy humans. All three participants independently exhibited robust ~12-h transcriptional rhythms in molecular programs involved in RNA and protein metabolism, with strong homology to circatidal gene programs previously identified in Cnidarian marine species.
Collapse
Affiliation(s)
- Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Silvia Liu
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Natalie L. David
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - William Dion
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Nandini K. Doshi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Lauren B. Siegel
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tânia Amorim
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Rosemary E. Andrews
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - G. V. Naveen Kumar
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Hanwen Li
- Department of Statistics, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA USA
| | - Saad Irfan
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tristan Pesaresi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Ankit X. Sharma
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Michelle Sun
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Pouneh K. Fazeli
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Matthew L. Steinhauser
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Sangoi AR, Tsai H, Harik L, Mahlow J, Tretiakova M, Williamson SR, Hirsch MS. Vascular, adipose tissue, and/or calyceal invasion in clear cell tubulopapillary renal cell tumour: potentially problematic diagnostic scenarios. Histopathology 2024; 84:1167-1177. [PMID: 38422612 DOI: 10.1111/his.15166] [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: 11/19/2023] [Revised: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024]
Abstract
AIMS The 2022 WHO classification for kidney tumours recently downgraded clear cell tubulopapillary (also known as clear cell papillary) renal cell carcinoma (RCC) to a benign neoplasm (i.e. clear cell tubulopapillary renal cell tumour) based on the overwhelmingly banal nature of this neoplasm. However, it has been recognized that some clear cell tubulopapillary renal cell tumours demonstrate vascular, adipose or pelvicalyceal invasion, raising the possibility of more aggressive behaviour. The goal of this study was to determine if these 'high stage' features have an effect on tumour prognosis, warranting a carcinoma designation. METHODS AND RESULTS After excluding cases with tissue artefact (i.e. prior core biopsy track changes) and other RCC subtypes with next-generation sequencing, nine clear cell tubulopapillary renal cell tumours with these so-called 'high stage' features, and otherwise classic morphologic and immunophenotypic findings, including low-grade cytology and 'cup-like' CA9 expression, were evaluated. Median tumour size was 2.2 cm with a range of 0.8 to 6.7 cm. Eight cases (89%) demonstrated perinephric or hilar adipose tissue invasion, although most of these cases showed a bulging (in contrast to an infiltrative) growth pattern. One case demonstrated renal vascular invasion in addition to hilar adipose tissue invasion, and one case demonstrated extension into the pelvicalyceal system. There were no recurrences or evidence of metastatic disease. CONCLUSION These overall findings continue to support the benign designation for clear cell tubulopapillary renal cell tumours, despite morphologic features that might raise the possibility of a 'higher stage' neoplasm.
Collapse
Affiliation(s)
- Ankur R Sangoi
- Department of Pathology, Stanford Medical Center, Stanford, CA, USA
| | - Harrison Tsai
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Center for Advanced Molecular Diagnostics, Brigham and Womens Hospital, Boston, MA, USA
| | - Lara Harik
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Jonathan Mahlow
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Coffey NJ, Simon MC. Metabolic alterations in hereditary and sporadic renal cell carcinoma. Nat Rev Nephrol 2024; 20:233-250. [PMID: 38253811 PMCID: PMC11165401 DOI: 10.1038/s41581-023-00800-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/24/2024]
Abstract
Kidney cancer is the seventh leading cause of cancer in the world, and its incidence is on the rise. Renal cell carcinoma (RCC) is the most common form and is a heterogeneous disease comprising three major subtypes that vary in their histology, clinical course and driver mutations. These subtypes include clear cell RCC, papillary RCC and chromophobe RCC. Molecular analyses of hereditary and sporadic forms of RCC have revealed that this complex and deadly disease is characterized by metabolic pathway alterations in cancer cells that lead to deregulated oxygen and nutrient sensing, as well as impaired tricarboxylic acid cycle activity. These metabolic changes facilitate tumour growth and survival. Specifically, studies of the metabolic features of RCC have led to the discovery of oncometabolites - fumarate and succinate - that can promote tumorigenesis, moonlighting functions of enzymes, and substrate auxotrophy owing to the disruption of pathways that enable the production of arginine and cholesterol. These metabolic alterations within RCC can be exploited to identify new therapeutic targets and interventions, in combination with novel approaches that minimize the systemic toxicity of metabolic inhibitors and reduce the risk of drug resistance owing to metabolic plasticity.
Collapse
Affiliation(s)
- Nathan J Coffey
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Zhang W, Zhang L, Wen Z, Liang J, Wang Y, Wang Z, Yin Z, Fan L. Clear-cell papillary renal cell tumour: New insights into clinicopathological features and molecular landscape after renaming by 5th WHO classification. Pathol Res Pract 2024; 255:155167. [PMID: 38324963 DOI: 10.1016/j.prp.2024.155167] [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: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVE Clear cell papillary renal cell tumour (CCPRCT) is a kind of renal epithelial cell tumor, and was renamed by the 5th WHO due to its specific epidemiology and clinicopathological characteristics. However, the biological mechanism and molecular basis of CCPRCT still need to be further clarified. This study aims to comprehensively evaluate clinicopathologic and molecular characteristics of CCPRCC, and particularly compare it with other more prevalent subtypes of renal cell carcinoma. METHODS 12 cases of CCPRCT were collected for analyzing the clinicopathological characteristics. Then, whole-exome sequencing (WES) was employed to reveal the genetic profiles, followed by comparison with the molecular genetic alterations identified in ccRCC (341) and pRCC (200) datasets obtained from the TCGA database. RESULTS Of the 12 CCPRCT cases, the male-to-female ratio was 4:1 with a mean age of 49.5 years (48.5 ± 10.5) at diagnosis. All patients were diagnosed accidentally during routine physical examinations. All tumors (12/12, 100%)had a solid-cystic appearance with a well-defined fibrous capsule. The median size of the tumors was 3 cm (2.98 ± 1.2). Histologically, the cystic papillary structures were considered to be prominent, lined with cuboidal tumor cells away from basement membrane. The tumor cells were moderately atypia equivalent to grade 1 or grade 2 according to the ISUP nuclear grading system. Typically, the tumor cell diffusely positive for CK7 and CAIX in a "cup-like" pattern. The results of WES revealed recurrent gene alterations (mainly missense mutation) of TTN and FLT in 4 cases (4/12, 33.3%), respectively, of which, the alteration of FLT was not observed in ccRCC and pRCC of the TCGA database. Other gene alterations including POTEC (1 cases), PRADC1 (1 cases), ZZZ3 (1 case) and PTPRZ1 (1 case), etc. Moreover, all of the CCPRCT cases displayed a lower tumor mutation burden (TMB) compared to ccRCC and pRCC with median TMB of 1.04 (range: 1.94 ± 2.74). None of the patients experienced tumor metastasis, recurrence, or tumor-related deaths. CONCLUSION CCPRCT is a renal epithelial cell tumor characterized by specific clinical and pathological features. Our study provides additional evidence supporting the favorable prognosis of CCPRCT. Furthermore, the potential molecular alterations were uncovered by this study in CCPRCT such as the FLT family and TTN. However, due to the limited sample size, larger studies are required to validate these findings.
Collapse
Affiliation(s)
- Wenhui Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Zhang
- Department of Pathology, The First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Zhu Wen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jiayi Liang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yingmei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhe Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Zhiyong Yin
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Linni Fan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, School of Basic Medicine and Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
14
|
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
|
15
|
Pei J, Zhang J, Cong Q. Computational analysis of protein-protein interactions of cancer drivers in renal cell carcinoma. FEBS Open Bio 2024; 14:112-126. [PMID: 37964489 PMCID: PMC10761929 DOI: 10.1002/2211-5463.13732] [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: 06/16/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023] Open
Abstract
Renal cell carcinoma (RCC) is the most common type of kidney cancer with rising cases in recent years. Extensive research has identified various cancer driver proteins associated with different subtypes of RCC. Most RCC drivers are encoded by tumor suppressor genes and exhibit enrichment in functional categories such as protein degradation, chromatin remodeling, and transcription. To further our understanding of RCC, we utilized powerful deep-learning methods based on AlphaFold to predict protein-protein interactions (PPIs) involving RCC drivers. We predicted high-confidence complexes formed by various RCC drivers, including TCEB1, KMT2C/D and KDM6A of the COMPASS-related complexes, TSC1 of the MTOR pathway, and TRRAP. These predictions provide valuable structural insights into the interaction interfaces, some of which are promising targets for cancer drug design, such as the NRF2-MAFK interface. Cancer somatic missense mutations from large datasets of genome sequencing of RCCs were mapped to the interfaces of predicted and experimental structures of PPIs involving RCC drivers, and their effects on the binding affinity were evaluated. We observed more than 100 cancer somatic mutations affecting the binding affinity of complexes formed by key RCC drivers such as VHL and TCEB1. These findings emphasize the importance of these mutations in RCC pathogenesis and potentially offer new avenues for targeted therapies.
Collapse
Affiliation(s)
- Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Jing Zhang
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
16
|
Liu Y, Li Y, Xu H, Zhou L, Yang X, Wang C. Exploration of Morphological Features of Clear Cell Renal Cell Carcinoma With PBRM1, SETD2, BAP1, or KDM5C Mutations. Int J Surg Pathol 2023; 31:1485-1494. [PMID: 36911986 DOI: 10.1177/10668969231157317] [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: 03/14/2023]
Abstract
The last decade has seen great advances in genomic profiling and prognosis-associated factors of clear cell renal cell carcinoma (RCC), the most common entity in kidney cancer. Following VHL, PBRM1, SETD2, BAP1, and KDM5C have been validated as the most common co-occurring gene mutations in clear cell RCC by multicenter studies. However, the morphological features of clear cell RCC with co-occurring gene mutations remain unclear. In this study, we presented 20 clear cell RCCs that underwent next-generation sequencing, of which 1 tumor was reclassified as ELOC-mutated RCC. PBRM1, SETD2, BAP1, and KDM5C were the most common mutations, following VHL. Morphologically, clear cell RCC with PBRM1 or KDM5C mutation usually displayed a low-grade pattern. Cystic changes and hyalinized stroma were often observed. The Ki67 index was <10%. These observations indicated good prognosis. However, mutated SETD2 may increase the malignancy of clear cell RCC with PBRM1 mutation. Two clear cell RCCs with mutated PBRM1 and SETD2 developed local or distant metastases. Clear cell RCC with BAP1 mutations always had high-grade patterns, and rhabdoid differentiation was also observed, indicating that BAP1 mutation was associated with poor outcomes. Papillary architecture was often a feature of BAP1 mutation, which is uncommon in clear cell RCC. PDL1 was positive in only one tumor with BAP1 mutation, and the positivity rate was limited to 5%. B7H3 was negative in all tumors. Morphologic findings in this small cohort may suggest why PBRM1 mutation does not correlate with decreased survival, whereas BAP1 mutation usually predicts poor outcomes.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunhao Li
- The First Clinical College, China Medical University, Shenyang, China
| | - Haimin Xu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Luting Zhou
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqun Yang
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaofu Wang
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
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: 13] [Impact Index Per Article: 6.5] [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
|
18
|
Sharma D, Xu W. ReGeNNe: genetic pathway-based deep neural network using canonical correlation regularizer for disease prediction. Bioinformatics 2023; 39:btad679. [PMID: 37963055 PMCID: PMC10666205 DOI: 10.1093/bioinformatics/btad679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 11/16/2023] Open
Abstract
MOTIVATION Common human diseases result from the interplay of genes and their biologically associated pathways. Genetic pathway analyses provide more biological insight as compared to conventional gene-based analysis. In this article, we propose a framework combining genetic data into pathway structure and using an ensemble of convolutional neural networks (CNNs) along with a Canonical Correlation Regularizer layer for comprehensive prediction of disease risk. The novelty of our approach lies in our two-step framework: (i) utilizing the CNN's effectiveness to extract the complex gene associations within individual genetic pathways and (ii) fusing features from ensemble of CNNs through Canonical Correlation Regularization layer to incorporate the interactions between pathways which share common genes. During prediction, we also address the important issues of interpretability of neural network models, and identifying the pathways and genes playing an important role in prediction. RESULTS Implementation of our methodology into three real cancer genetic datasets for different prediction tasks validates our model's generalizability and robustness. Comparing with conventional models, our methodology provides consistently better performance with AUC improvement of 11% on predicting early/late-stage kidney cancer, 10% on predicting kidney versus liver cancer type and 7% on predicting survival status in ovarian cancer as compared to the next best conventional machine learning model. The robust performance of our deep learning algorithm indicates that disease prediction using neural networks in multiple functionally related genes across different pathways improves genetic data-based prediction and understanding molecular mechanisms of diseases. AVAILABILITY AND IMPLEMENTATION https://github.com/divya031090/ReGeNNe.
Collapse
Affiliation(s)
- Divya Sharma
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G2C4, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Wei Xu
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G2C4, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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: 12] [Impact Index Per Article: 6.0] [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
|
24
|
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
|
25
|
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: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [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
|
26
|
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
|
27
|
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
|
28
|
Taylor AS, Skala SL. Tumors masquerading as type 2 papillary renal cell carcinoma: pathologists' ever-expanding differential diagnosis for a heterogeneous group of entities. Urol Oncol 2022; 40:499-511. [PMID: 34116938 DOI: 10.1016/j.urolonc.2021.04.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/07/2023]
Abstract
Although papillary renal cell carcinoma has historically been classified as either type 1 or type 2, data from The Cancer Genome Atlas (TCGA) has demonstrated significant genomic heterogeneity in tumors classified as "type 2 papillary renal cell carcinoma" (T2PRCC). Papillary renal cell carcinoma is expected to have a favorable clinical course compared to clear cell renal cell carcinoma (CCRCC). However, tumors with poor outcome more similar to CCRCC were included in the T2PRCC cohort studied by the TCGA. The differential diagnosis for T2PRCC includes a variety of other renal tumors, including aggressive entities such as TFE3 translocation-associated renal cell carcinoma, TFEB-amplified renal cell carcinoma, fumarate hydratase-deficient renal cell carcinoma, high-grade CCRCC, and collecting duct carcinoma. Accurate classification of these tumors is important for prognostication and selection of therapy.
Collapse
Affiliation(s)
- Alexander S Taylor
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - Stephanie L Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI.
| |
Collapse
|
29
|
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
|
30
|
Contemporary Clinical Definitions, Differential Diagnosis, and Novel Predictive Tools for Renal Cell Carcinoma. Biomedicines 2022; 10:biomedicines10112926. [PMID: 36428491 PMCID: PMC9687297 DOI: 10.3390/biomedicines10112926] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/26/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant progress regarding clinical detection/imaging evaluation modalities and genetic/molecular characterization of pathogenesis, advanced renal cell carcinoma (RCC) remains an incurable disease and overall RCC mortality has been steadily rising for decades. Concomitantly, clinical definitions have been greatly nuanced and refined. RCCs are currently viewed as a heterogeneous series of cancers, with the same anatomical origin, but fundamentally different metabolisms and clinical behaviors. Thus, RCC pathological diagnosis/subtyping guidelines have become increasingly intricate and cumbersome, routinely requiring ancillary studies, mainly immunohistochemistry. Meanwhile, RCC-associated-antigen targeted systemic therapy has been greatly diversified and emerging, novel clinical applications for RCC immunotherapy have already reported significant survival benefits, at least in the adjuvant setting. Even so, systemically disseminated RCCs still associate very poor clinical outcomes, with currently available therapeutic modalities only being able to prolong survival. In lack of a definitive cure for advanced RCCs, integration of the amounting scientific knowledge regarding RCC pathogenesis into RCC clinical management has been paramount for improving patient outcomes. The current review aims to offer an integrative perspective regarding contemporary RCC clinical definitions, proper RCC clinical work-up at initial diagnosis (semiology and multimodal imaging), RCC pathological evaluation, differential diagnosis/subtyping protocols, and novel clinical tools for RCC screening, risk stratification and therapeutic response prediction.
Collapse
|
31
|
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
|
32
|
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: 330] [Impact Index Per Article: 110.0] [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
|
33
|
Nero C, Boldrini L, Lenkowicz J, Giudice MT, Piermattei A, Inzani F, Pasciuto T, Minucci A, Fagotti A, Zannoni G, Valentini V, Scambia G. Deep-Learning to Predict BRCA Mutation and Survival from Digital H&E Slides of Epithelial Ovarian Cancer. Int J Mol Sci 2022; 23:ijms231911326. [PMID: 36232628 PMCID: PMC9570450 DOI: 10.3390/ijms231911326] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
BRCA 1/2 genes mutation status can already determine the therapeutic algorithm of high grade serous ovarian cancer patients. Nevertheless, its assessment is not sufficient to identify all patients with genomic instability, since BRCA 1/2 mutations are only the most well-known mechanisms of homologous recombination deficiency (HR-d) pathway, and patients displaying HR-d behave similarly to BRCA mutated patients. HRd assessment can be challenging and is progressively overcoming BRCA testing not only for prognostic information but more importantly for drugs prescriptions. However, HR testing is not already integrated in clinical practice, it is quite expensive and it is not refundable in many countries. Selecting patients who are more likely to benefit from this assessment (BRCA 1/2 WT patients) at an early stage of the diagnostic process, would allow an optimization of genomic profiling resources. In this study, we sought to explore whether somatic BRCA1/2 genes status can be predicted using computational pathology from standard hematoxylin and eosin histology. In detail, we adopted a publicly available, deep-learning-based weakly supervised method that uses attention-based learning to automatically identify sub regions of high diagnostic value to accurately classify the whole slide (CLAM). The same model was also tested for progression free survival (PFS) prediction. The model was tested on a cohort of 664 (training set: n = 464, testing set: n = 132) ovarian cancer patients, of whom 233 (35.1%) had a somatic BRCA 1/2 mutation. An area under the curve of 0.7 and 0.55 was achieved in the training and testing set respectively. The model was then further refined by manually identifying areas of interest in half of the cases. 198 images were used for training (126/72) and 87 images for validation (55/32). The model reached a zero classification error on the training set, but the performance was 0.59 in terms of validation ROC AUC, with a 0.57 validation accuracy. Finally, when applied to predict PFS, the model achieved an AUC of 0.71, with a negative predictive value of 0.69, and a positive predictive value of 0.75. Based on these analyses, we have planned further steps of development such as proving a reference classification performance, exploring the hyperparameters space for training optimization, eventually tweaking the learning algorithms and the neural networks architecture for better suiting this specific task. These actions may allow the model to improve performances for all the considered outcomes.
Collapse
Affiliation(s)
- Camilla Nero
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-30154979
| | - Luca Boldrini
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiomics Core Facility, 00168 Rome, Italy
| | - Jacopo Lenkowicz
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiomics Core Facility, 00168 Rome, Italy
| | - Maria Teresa Giudice
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| | - Alessia Piermattei
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Frediano Inzani
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Tina Pasciuto
- Fondazione Policlinico Agostino Gemelli, IRCCS, Data Collection Core Facility, 00168 Rome, Italy
| | - Angelo Minucci
- Fondazione Policlinico Agostino Gemelli, IRCCS, Genomics Core Facility, 00168 Rome, Italy
| | - Anna Fagotti
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| | - Gianfranco Zannoni
- Fondazione Policlinico Agostino Gemelli, IRCCS, Pathology, 00168 Rome, Italy
| | - Vincenzo Valentini
- Fondazione Policlinico Agostino Gemelli, IRCCS, Radiation Oncology, 00168 Rome, Italy
| | - Giovanni Scambia
- Fondazione Policlinico Agostino Gemelli, IRCCS, Gynecology and Obstetrics, 00168 Rome, Italy
| |
Collapse
|
34
|
Mohanty SK, Lobo A, Cheng L. The 2022 revision of World Health Organization classification of tumors of the urinary system and male genital organs: advances and challenges. Hum Pathol 2022; 136:123-143. [PMID: 36084769 DOI: 10.1016/j.humpath.2022.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 01/07/2023]
Abstract
The fifth edition of the World Health Organization (WHO) classification of urinary and male genital organ tumors has been recently published in 2022. The application of molecular profiling has made a substantial impact on classification of urologic tumors. The new WHO classification introduces a group of molecularly well-defined renal tumor subtypes. The significant changes include addition of a category of "other oncocytic tumors" with oncocytoma/chromophobe renal cell carcinoma (chRCC)-like features, elimination of the subcategorization of type1/2 papillary RCC and inclusion of eosinophilic solid and cystic RCC as an independent tumor entity. The WHO/ISUP grading now has been recommended for all RCCs. Major nomenclature changes include replacement of histologic 'variants' by 'subtypes', 'clear cell papillary renal cell carcinoma' to 'clear cell renal cell tumor','TCEB1-mutated RCC' to 'ELOC-mutated RCC', 'hereditary leiomyomatosis and renal cell carcinoma' to 'fumarate hydratase-deficient RCC', 'RCC-Unclassified' to 'RCC-NOS', 'primitive neuroectodermal tumor' to 'embryonic neuroectodermal tumor', 'testicular carcinoid' to 'testicular neuroendocrine tumor', and 'basal cell carcinoma of the prostate' to 'adenoid-cystic (basal-cell) carcinoma of the prostate'. Metastatic, hematolymphoid, mesenchymal, melanocytic, soft tissue and neuroendocrine tumors are collectively discussed in separate chapters. It has been suggested that the morphological classification of urothelial cancer be replaced with a new molecular taxonomic classification system.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute and CORE Diagnostics, Gurgaon, India (Zipcode:122016)
| | - Anandi Lobo
- Department of Pathology and Laboratory Medicine, Kapoor Center of Urology and Pathology, Raipur, India (Zipcode:490042)
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Lifespan Academic Medical Center, Providence, RI, USA (Zipcode: 02903).
| |
Collapse
|
35
|
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
|
36
|
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]
|
37
|
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
|
38
|
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
|
39
|
Evaluating Established Roles, Future Perspectives and Methodological Heterogeneity for Wilms’ Tumor 1 (WT1) Antigen Detection in Adult Renal Cell Carcinoma, Using a Novel N-Terminus Targeted Antibody (Clone WT49). Biomedicines 2022; 10:biomedicines10040912. [PMID: 35453662 PMCID: PMC9026801 DOI: 10.3390/biomedicines10040912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 11/20/2022] Open
Abstract
Renal cell carcinoma (RCC) is arguably the deadliest form of genitourinary malignancy and is nowadays viewed as a heterogeneous series of cancers, with the same origin but fundamentally different metabolisms and clinical behaviors. Immunohistochemistry (IHC) is increasingly necessary for RCC subtyping and definitive diagnosis. WT1 is a complex gene involved in carcinogenesis. To address reporting heterogeneity and WT1 IHC standardization, we used a recent N-terminus targeted monoclonal antibody (clone WT49) to evaluate WT1 protein expression in 56 adult RCC (aRCC) cases. This is the largest WT1 IHC investigation focusing exclusively on aRCCs and the first report on clone WT49 staining in aRCCs. We found seven (12.5%) positive cases, all clear cell RCCs, showing exclusively nuclear staining for WT1. We did not disregard cytoplasmic staining in any of the negative cases. Extratumoral fibroblasts, connecting tubules and intratumoral endothelial cells showed the same exclusively nuclear WT1 staining pattern. We reviewed WT1 expression patterns in aRCCs and the possible explanatory underlying metabolomics. For now, WT1 protein expression in aRCCs is insufficiently investigated, with significant discrepancies in the little data reported. Emerging WT1-targeted RCC immunotherapy will require adequate case selection and sustained efforts to standardize the quantification of tumor-associated antigens for aRCC and its many subtypes.
Collapse
|
40
|
Wang Z, Zeng Z, Starkuviene V, Erfle H, Kan K, Zhang J, Gunkel M, Sticht C, Rahbari N, Keese M. MicroRNAs Influence the Migratory Ability of Human Umbilical Vein Endothelial Cells. Genes (Basel) 2022; 13:genes13040640. [PMID: 35456446 PMCID: PMC9029696 DOI: 10.3390/genes13040640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
To identify miRNAs that are involved in cell migration in human umbilical vein endothelial cells (HUVECs), we employed RNA sequencing under high glucose incubation and text mining within the databases miRWalk and TargetScanHuman using 83 genes that regulate HUVECs migration. From both databases, 307 predicted miRNAs were retrieved. Differentially expressed miRNAs were determined by exposing HUVECs to high glucose stimulation, which significantly inhibited the migratory ability of HUVECs as compared to cells cultured in normal glucose. A total of 35 miRNAs were found as differently expressed miRNAs in miRNA sequencing, and 4 miRNAs, namely miR-21-3p, miR-107, miR-143-3p, and miR-106b-5p, were identified as overlapping hits. These were subjected to hub gene analysis and pathway analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG), identifing 71 pathways which were influenced by all four miRNAs. The influence of all four miRNAs on HUVEC migration was phenomorphologically confirmed. miR21 and miR107 promoted migration in HUVECs while miR106b and miR143 inhibited migration. Pathway analysis also revealed eight shared pathways between the four miRNAs. Protein–protein interaction (PPI) network analysis was then performed to predict the functionality of interacting genes or proteins. This revealed six hub genes which could firstly be predicted to be related to HUVEC migration.
Collapse
Affiliation(s)
- Zhaohui Wang
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Ziwei Zeng
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Vytaute Starkuviene
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
- Institute of Biosciences, Vilnius University Life Sciences Center, 10257 Vilnius, Lithuania
- Correspondence: (V.S.); (M.K.)
| | - Holger Erfle
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Kejia Kan
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Jian Zhang
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Manuel Gunkel
- BioQuant, Heidelberg University, 69120 Heidelberg, Germany; (H.E.); (M.G.)
| | - Carsten Sticht
- NGS Core Facility, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Nuh Rahbari
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
| | - Michael Keese
- Vascular Surgery, University Clinic Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (Z.W.); (Z.Z.); (K.K.); (J.Z.); (N.R.)
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Correspondence: (V.S.); (M.K.)
| |
Collapse
|
41
|
Andreou A, Yngvadottir B, Bassaganyas L, Clark G, Martin E, Whitworth J, Cornish AJ, Genomics England Research Consortium, Houlston RS, Rich P, Egan C, Hodgson SV, Warren AY, Snape K, Maher ER. Elongin C (ELOC/TCEB1)-associated von Hippel-Lindau disease. Hum Mol Genet 2022; 31:2728-2737. [PMID: 35323939 PMCID: PMC9402235 DOI: 10.1093/hmg/ddac066] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 12/02/2022] Open
Abstract
Around 95% of patients with clinical features that meet the diagnostic criteria for von Hippel-Lindau disease (VHL) have a detectable inactivating germline variant in VHL. The VHL protein (pVHL) functions as part of the E3 ubiquitin ligase complex comprising pVHL, elongin C, elongin B, cullin 2 and ring box 1 (VCB-CR complex), which plays a key role in oxygen sensing and degradation of hypoxia-inducible factors. To date, only variants in VHL have been shown to cause VHL disease. We undertook trio analysis by whole-exome sequencing in a proband with VHL disease but without a detectable VHL mutation. Molecular studies were also performed on paired DNA extracted from the proband's kidney tumour and blood and bioinformatics analysis of sporadic renal cell carcinoma (RCC) dataset was undertaken. A de novo pathogenic variant in ELOC NM_005648.4(ELOC):c.236A>G (p.Tyr79Cys) gene was identified in the proband. ELOC encodes elongin C, a key component [C] of the VCB-CR complex. The p.Tyr79Cys substitution is a mutational hotspot in sporadic VHL-competent RCC and has previously been shown to mimic the effects of pVHL deficiency on hypoxic signalling. Analysis of an RCC from the proband showed similar findings to that in somatically ELOC-mutated RCC (expression of hypoxia-responsive proteins, no somatic VHL variants and chromosome 8 loss). These findings are consistent with pathogenic ELOC variants being a novel cause for VHL disease and suggest that genetic testing for ELOC variants should be performed in individuals with suspected VHL disease with no detectable VHL variant.
Collapse
Affiliation(s)
- Avgi Andreou
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Bryndis Yngvadottir
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Laia Bassaganyas
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Graeme Clark
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK,Stratified Medicine Core Laboratory NGS Hub, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Ezequiel Martin
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK,Stratified Medicine Core Laboratory NGS Hub, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - James Whitworth
- Department of Medical Genetics, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | | | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, UK
| | - Philip Rich
- Department of Neuroradiology, St. George’s University Hospitals NHS Foundation Trust, London SW17 0QT, UK
| | - Catherine Egan
- NIHR Biomedical Research Center at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Shirley V Hodgson
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Anne Y Warren
- Department of Histopathology, Cambridge University NHS Foundation Trust, Cambridge CB2 OQQ, UK
| | - Katie Snape
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK,St George's University of London, UK
| | - Eamonn R Maher
- To whom correspondence should be addressed at: Department of Medical Genetics, University of Cambridge, Box 238, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK. Tel: +44 01223746715; Fax: +44 01223746777;
| |
Collapse
|
42
|
El-Zaatari ZM, Truong LD. Renal Cell Carcinoma in End-Stage Renal Disease: A Review and Update. Biomedicines 2022; 10:biomedicines10030657. [PMID: 35327459 PMCID: PMC8944945 DOI: 10.3390/biomedicines10030657] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022] Open
Abstract
Renal cell carcinoma (RCC) occurring in the setting of end-stage renal disease (ESRD) shows unique clinicopathological characteristics. The two most frequent types of ESRD-associated RCC are acquired cystic kidney disease-associated renal cell carcinoma (ACKD-RCC) and clear-cell papillary renal cell carcinoma (ccpRCC). Other types of RCC also occur in ESRD, albeit with different frequencies from the non-ESRD general population. The histological features of RCC do not vary in the setting of ESRD vs. non-ESRD, yet other findings, such as multifocality and multiple tumor types, are more frequent in ESRD. Studies have generated novel and important knowledge of the etiology, epidemiology, diagnosis, treatment, immunophenotype, and molecular characteristics of ESRD-associated RCC. Knowledge of these data is important for both pathologists and other physicians who may encounter ESRD patients with RCC. This review presents a comprehensive summary and update of the literature on RCC in ESRD, with a focus on the two most frequent types, ACKD-RCC and ccpRCC.
Collapse
Affiliation(s)
- Ziad M. El-Zaatari
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Main Building, Houston, TX 77030, USA;
- Weil Medical College, Cornell University, New York, NY 10022, USA
- Correspondence: ; Tel.: +1-713-441-6478; Fax: +1-713-793-1603
| | - Luan D. Truong
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Main Building, Houston, TX 77030, USA;
- Weil Medical College, Cornell University, New York, NY 10022, USA
| |
Collapse
|
43
|
Argani P, Mehra R. Renal cell carcinoma associated with tuberous sclerosis complex (TSC)/mammalian target of rapamycin (MTOR) genetic alterations. Mod Pathol 2022; 35:296-297. [PMID: 35046523 DOI: 10.1038/s41379-021-00971-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/04/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023]
Affiliation(s)
- Pedram Argani
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
44
|
Alaghehbandan R, Siadat F, Trpkov K. What's new in the WHO 2022 classification of kidney tumours? Pathologica 2022; 115:8-22. [PMID: 36645398 DOI: 10.32074/1591-951x-818] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 01/17/2023] Open
Abstract
The World Health Organization (WHO) 2022 classification of urinary and male genital tumours (5th edition) has significantly improved our understanding of the morphologic, immunohistochemical, and molecular characteristics of renal tumours. The aim of this review is to outline the most important changes and diagnostic updates in the WHO 2022 classification of kidney tumours. A major change in this edition is the grouping of renal tumours into broader categories that include "clear cell renal tumours", "papillary renal tumours", "oncocytic and chromophobe renal tumours", "collecting duct tumours" as well as adding two categories of "other renal tumours" and "molecularly defined renal carcinomas". Novel entities included in the WHO 2022 classification are eosinophilic solid and cystic renal cell carcinoma (ESC RCC), anaplastic lymphoma kinase (ALK)-rearranged RCC and ELOC (formerly TCEB1)-mutated RCC. The category of "other renal tumours" includes a group of diverse, unrelated renal tumours that do not fit into other categories. The group of "molecularly defined renal carcinomas" reflects recent discoveries in the renal tumour genomics. These molecularly-defined renal entities demonstrate a set of morphologic features reflecting genotype-phenotype relationships. Final diagnosis of such entities rests on phenotypic and immunohistochemical (IHC) correlation, usually associated with IHC surrogate makers that reflect specific genetic abnormalities.
Collapse
Affiliation(s)
- Reza Alaghehbandan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Farshid Siadat
- Cumming School of Medicine, University of Calgary and Alberta Precision Laboratories, Calgary, AB, Canada
| | - Kiril Trpkov
- Cumming School of Medicine, University of Calgary and Alberta Precision Laboratories, Calgary, AB, Canada
| |
Collapse
|
45
|
Prochazkova K, Ptakova N, Alaghehbandan R, Williamson SR, Vaněček T, Vodicka J, Treska V, Rogala J, Pivovarcikova K, Michalova K, Slisarenko M, Hora M, Michal M, Hes O. Mutation Profile Variability in the Primary Tumor and Multiple Pulmonary Metastases of Clear Cell Renal Cell Carcinoma. A Review of the Literature and Analysis of Four Metastatic Cases. Cancers (Basel) 2021; 13:5906. [PMID: 34885018 PMCID: PMC8656868 DOI: 10.3390/cancers13235906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/13/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background: There are limited data concerning inter-tumoral and inter-metastatic heterogeneity in clear cell renal cell carcinoma (CCRCC). The aim of our study was to review published data and to examine mutation profile variability in primary and multiple pulmonary metastases (PMs) in our cohort of four patients with metastatic CCRCC. (2) Methods: Four patients were enrolled in this study. The clinical characteristics, types of surgeries, histopathologic results, immunohistochemical and genetic evaluations of corresponding primary tumor and PMs, and follow-up data were recorded. (3) Results: In our series, the most commonly mutated genes were those in the canonically dysregulated VHL pathway, which were detected in both primary tumors and corresponding metastasis. There were genetic profile differences between primary and metastatic tumors, as well as among particular metastases in one patient. (4) Conclusions: CCRCC shows heterogeneity between the primary tumor and its metastasis. Such mutational changes may be responsible for suboptimal treatment outcomes in targeted therapy settings.
Collapse
Affiliation(s)
- Kristyna Prochazkova
- Department of Surgery, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 304 60 Pilsen, Czech Republic; (K.P.); (J.V.); (V.T.)
| | - Nikola Ptakova
- Second Faculty of Medicine, Charles University, 150 06 Prague, Czech Republic;
| | - Reza Alaghehbandan
- Department of Pathology, University of British Columbia, Vancouver, BC 2329, Canada;
| | - Sean R. Williamson
- Robert J. Tomsich Pathology and Laboratory Medicine Institute and Glickman Urological Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Tomáš Vaněček
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Josef Vodicka
- Department of Surgery, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 304 60 Pilsen, Czech Republic; (K.P.); (J.V.); (V.T.)
| | - Vladislav Treska
- Department of Surgery, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 304 60 Pilsen, Czech Republic; (K.P.); (J.V.); (V.T.)
| | - Joanna Rogala
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Kristyna Pivovarcikova
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Kvetoslava Michalova
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Maryna Slisarenko
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Milan Hora
- Department of Urology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic;
| | - Michal Michal
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| | - Ondrej Hes
- Department of Pathology, Faculty of Medicine in Pilsen and University Hospital Pilsen, Charles University, 305 99 Pilsen, Czech Republic; (T.V.); (J.R.); (K.P.); (K.M.); (M.S.); (M.M.)
| |
Collapse
|
46
|
[Renal carcinoma with leiomyomatous stroma and osseous metaplasia from a patient diagnosed with a tuberous sclerosis complex]. Ann Pathol 2021; 41:557-560. [PMID: 34629215 DOI: 10.1016/j.annpat.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/22/2021] [Accepted: 06/14/2021] [Indexed: 11/24/2022]
Abstract
Renal cell carcinoma with leiomyomatous stroma is a rare and poorly described histopathological entity. Here we report a unique case with osseous metaplasia, in a 31-year-old man recently diagnosed with a tuberous sclerosis complex (TSC2 gene mutation). Partial nephrectomy was performed. Histologically, the epithelial component was made up of papillary and alveolar structures with clear to eosinophilic cytoplasm, and basally located nuclei. The cells are surrounded by an abundant smooth muscle stroma with focally osseous metaplasia. The tumor was positive for carbonic anhydrase IX, cytokeratin 7, cytokeratin 20, and CD10, and negative for TFE3. This emerging entity is highly correlated to tuberous sclerosis complex, which justifies a screening for the syndrome when this diagnosis is made.
Collapse
|
47
|
Lobo J, Ohashi R, Helmchen BM, Rupp NJ, Rüschoff JH, Moch H. The Morphological Spectrum of Papillary Renal Cell Carcinoma and Prevalence of Provisional/Emerging Renal Tumor Entities with Papillary Growth. Biomedicines 2021; 9:1418. [PMID: 34680535 PMCID: PMC8533532 DOI: 10.3390/biomedicines9101418] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Renal cell carcinoma (RCC) represents a heterogeneous disease, encompassing an increasing number of tumor subtypes. Post-2016, the World Health Organization (WHO) classification recognized that the spectrum of papillary renal cell carcinoma is evolving and has long surpassed the dichotomic simplistic "type 1 versus type 2" classification. The differential diagnosis of pRCC includes several new provisional/emerging entities with papillary growth. Type 2 tumors have been cleared out of several confounding entities, now regarded as independent tumors with specific clinical and molecular backgrounds. In this work we describe the prevalence and characteristics of emerging papillary tumor entities in two renal tumor cohorts (one consisting of consecutive papillary tumors from a single institute, the other consisting of consultation cases from several centers). After a review of 154 consecutive pRCC cases, 58% remained type 1 pRCC, and 34% type 2 pRCC. Papillary renal neoplasm with reversed polarity (1.3%), biphasic hyalinizing psammomatous RCC (1.3%), and biphasic squamoid/alveolar RCC (4.5%) were rare. Among 281 consultation cases, 121 (43%) tumors had a dominant papillary growth (most frequently MiT family translocation RCCs, mucinous tubular and spindle cell carcinoma and clear cell papillary RCC). Our data confirm that the spectrum of RCCs with papillary growth represents a major diagnostical challenge, frequently requiring a second expert opinion. Papillary renal neoplasm with reversed polarity, biphasic hyalinizing psammomatous RCC, and biphasic squamoid/alveolar RCC are rarely sent out for a second opinion, but correct classification and knowledge of these variants will improve our understanding of the clinical behavior of renal tumors with papillary growth.
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, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS—School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Riuko Ohashi
- Histopathology Core Facility, Faculty of Medicine, Niigata University, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan;
- Division of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, 1-757 Asahimachi-Dori, Chuo-Ku, Niigata 951-8510, Japan
| | - Birgit M. Helmchen
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Niels J. Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Jan H. Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; (B.M.H.); (N.J.R.); (J.H.R.)
- Faculty of Medicine, University of Zurich, Rämistrasse 71, 8006 Zurich, Switzerland
| |
Collapse
|
48
|
Tian X, Xu WH, Wu JL, Gan HL, Wang HK, Gu WJ, Qu YY, Zhang HL, Ye DW. Clear Cell Papillary Renal Cell Carcinoma Shares Distinct Molecular Characteristics and may be Significantly Associated With Higher Risk of Developing Second Primary Malignancy. Pathol Oncol Res 2021; 27:1609809. [PMID: 34512202 PMCID: PMC8432294 DOI: 10.3389/pore.2021.1609809] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
Traditionally, clear cell papillary renal cell carcinoma (ccpRCC) was considered to share similar molecular and histological characteristics with clear cell renal cell carcinoma (ccRCC) and papillary renal cell carcinoma (pRCC). Here we aimed to identify somatic and germline variants of ccpRCC. For this purpose, we conducted whole-exome sequencing to detect somatic variants in the tissues of 18 patients with pathologically confirmed ccpRCC, who underwent surgical treatment at Fudan University Shanghai Cancer Center. Targeted sequencing was conducted to detect germline variants in paired tumor or normal tissues or blood. Somatic and germline variants of ccRCC and Renal cell carcinoma included in The Cancer Genome Atlas data and other published data were analyzed as well. The molecular profiles of ccpRCC, ccRCC and pRCC were compared. Among the 387 somatic variants identified, TCEB1 (3/18) and VHL (3/18) variants occurred at the highest frequencies. Germline mutation detection showed that nine variants associated with Fanconi anemia (VAFAs) pathway (FANCA, 6/18; FANCI, 3/18) were identified in 18 ccpRCC patients. Among ccpRCC patients with VAFAs, five out of eight patients had second primary malignancy or family history of cancer. Somatic variants characteristics may distinguish ccpRCC from ccRCC or pRCC and germline VAFAs may be a molecular characterization of ccpRCC. Compared with ccRCC or pRCC, ccpRCC patients may be significantly correlated with higher risk of developing second primary malignancy.
Collapse
Affiliation(s)
- Xi Tian
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Hao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jun-Long Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hua-Lei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hong-Kai Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei-Jie Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan-Yuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hai-Liang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ding-Wei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Bergmann L, Weber S, Hartmann A, Ahrens M. Pathology and systemic therapy of non-clear cell renal cell carcinoma: an overview. Expert Rev Anticancer Ther 2021; 21:1273-1286. [PMID: 34291700 DOI: 10.1080/14737140.2021.1959319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Non-clear cell renal cell carcinoma (nccRCC) represents a highly heterogenous group of kidney cancer entities. As most clinical trials predominantly include patients with clear cell RCC (ccRCC), nccRCC treatment guidelines are mainly extrapolated from recommendations in ccRCC. Here, we review and elucidate current data on the pathologic classification and treatment of nccRCC.Areas covered: This article gives an overview of the WHO classification of RCC, showing the histological diversity of nccRCC and focusing particularly on entities first characterized since 2016, their specific molecular behavior and their role as indicators for hereditary cancer syndromes. In this context, we discuss the available data on nccRCC treatment oprtions such as tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, cytotoxic chemotherapy, and immune checkpoint inhibitors.Expert opinion: Although nccRCCs are relatively uncommon, entities of this type account for a subgroup of up to 20-25% of all RCCs. Advances in histopathology and molecular genetics, together with evidence gained from retrospective and prospective clinical data, have improved understanding of these tumors in recent years. Nevertheless, selective trials of current and novel therapies including new targeted agents in patients with nccRCC are urgently needed to further improve treatment guidelines.
Collapse
Affiliation(s)
- Lothar Bergmann
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany.,Private Praxis for Hematology/Oncology, Schifferstrasse, Frankfurt, Germany
| | - Sarah Weber
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany
| | - Arndt Hartmann
- Institute for Pathology, University Hospital, Erlangen, Nürnberg, Germany
| | - Marit Ahrens
- Medical Clinic II, J.W. Goethe University, Frankfurt, Germany
| |
Collapse
|
50
|
Baniak N, Barletta JA, Hirsch MS. Key Renal Neoplasms With a Female Predominance. Adv Anat Pathol 2021; 28:228-250. [PMID: 34009777 DOI: 10.1097/pap.0000000000000301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Renal neoplasms largely favor male patients; however, there is a growing list of tumors that are more frequently diagnosed in females. These tumors include metanephric adenoma, mixed epithelial and stromal tumor, juxtaglomerular cell tumor, mucinous tubular and spindle cell carcinoma, Xp11.2 (TFE3) translocation-associated renal cell carcinoma, and tuberous sclerosis complex (somatic or germline) associated renal neoplasms. The latter category is a heterogenous group with entities still being delineated. Eosinophilic solid and cystic renal cell carcinoma is the best-described entity, whereas, eosinophilic vacuolated tumor is a proposed entity, and the remaining tumors are currently grouped together under the umbrella of tuberous sclerosis complex/mammalian target of rapamycin-related renal neoplasms. The entities described in this review are often diagnostic considerations when evaluating renal mass tissue on biopsy or resection. For example, Xp11.2 translocation renal cell carcinoma is in the differential when a tumor has clear cell cytology and papillary architecture and occurs in a young or middle-aged patient. In contrast, tuberous sclerosis complex-related neoplasms often enter the differential for tumors with eosinophilic cytology. This review provides an overview of the clinical, gross, microscopic, immunohistochemical, genetic, and molecular alterations in key renal neoplasms occurring more commonly in females; differential diagnoses are also discussed regardless of sex predilection.
Collapse
Affiliation(s)
- Nicholas Baniak
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Michelle S Hirsch
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|