1
|
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
|
2
|
Alaghehbandan R, Campbell SC, McKenney JK. Evolution in the Pathologic Classification of Renal Neoplasia. Urol Clin North Am 2023; 50:181-189. [PMID: 36948665 DOI: 10.1016/j.ucl.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
The pathologic classification of renal tumors is a dynamic and complex process, which has evolved to a "histomolecular" driven system. Despite advances in molecular characterization, most renal tumors can be diagnosed by morphology with or without using a limited set of immunohistochemical stains. If access to molecular resources and specific immunohistochemical markers is limited, pathologists may face difficulties in following an optimal algorithm to classify renal tumors. In this article, we detail the historical evolution of renal tumor classification, including a synopsis of major changes introduced by the current fifth edition World Health Organization 2022 classification of renal epithelial tumors.
Collapse
Affiliation(s)
- Reza Alaghehbandan
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 2119 E. 96th Street, L25, Cleveland, OH 44106, USA
| | - Steven C Campbell
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Q10-120, Glickman Tower, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Jesse K McKenney
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, 2119 E. 96th Street, L25, Cleveland, OH 44106, USA.
| |
Collapse
|
3
|
Lu SQ, Lv W, Liu YJ, Deng H. Fat-poor renal angiomyolipoma with prominent cystic degeneration: A case report and review of the literature. World J Clin Cases 2023; 11:417-425. [PMID: 36686346 PMCID: PMC9850960 DOI: 10.12998/wjcc.v11.i2.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/16/2022] [Accepted: 12/21/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Angiomyolipoma (AML), the most common benign tumor of the kidney, is usually composed of dysmorphic blood vessels, smooth muscle, and mature adipose tissue. To our knowledge, AML with cystic degeneration has rarely been documented. Cystic degeneration, hemorrhage, and a lack of fat bring great challenges to the diagnosis.
CASE SUMMARY A 60-year-old man with hypertension presented with a 5-year history of cystic mass in his left kidney. He fell 2 mo ago. A preoperative computed tomography (CT) scan showed a mixed-density cystic lesion without macroscopic fat density, the size of which had increased compared with before, probably due to hemorrhage caused by a trauma. Radical nephrectomy was performed. Histopathological studies revealed that the lesion mainly consisted of tortuous, ectatic, and thick-walled blood vessels, mature adipose tissue, and smooth muscle-like spindle cells arranged around the abnormal blood vessels. The tumor cells exhibited positivity for human melanoma black-45, Melan-A, smooth muscle actin, calponin, S-100, and neuron-specific enolase, rather than estrogen receptor, progesterone receptor, CD68, and cytokeratin. The Ki-67 labeling index was less than 5%. The final diagnosis was a fat-poor renal AML (RAML) with prominent cystic degeneration.
CONCLUSION When confronting a large renal cystic mass, RAML should be included in the differential diagnosis.
Collapse
Affiliation(s)
- Shi-Qi Lu
- Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Wei Lv
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - You-Jun Liu
- Department of Radiology, The Fourth Affiliated Hospital of Nanchang University, Nanchang 330003, Jiangxi Province, China
| | - Huan Deng
- Department of Pathology, The Fourth Affiliated Hospital of Nanchang University, Nanchang 330003, Jiangxi Province, China
| |
Collapse
|
4
|
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: 1.0] [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
|
5
|
Papillary renal cell carcinoma: a single institutional study of 199 cases addressing classification, clinicopathologic and molecular features, and treatment outcome. Mod Pathol 2022; 35:825-835. [PMID: 34949764 PMCID: PMC9177523 DOI: 10.1038/s41379-021-00990-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/24/2021] [Accepted: 11/27/2021] [Indexed: 01/20/2023]
Abstract
The morphologic spectrum of type 1 papillary renal cell carcinoma (PRCC) is not well-defined, since a significant proportion of cases have mixed type 1 and 2 histology. We analyzed 199 cases of PRCC with any (even if focal) type 1 features, with a median follow-up of 12 years, to identify clinicopathological features associated with outcome. Ninety-five tumors (48%) of the cohort contained some type 2 component (median amount: 25%; IQR: 10%, 70%). As a group they showed high rates of progression-free (PFS) and cancer-specific survival (CSS). Tumor size, mitotic rate, lymphovascular invasion, sarcomatoid differentiation, sheet-like architecture, and lack of tumor circumscription were significantly associated with CSS (p ≤ 0.015) on univariate analysis. While predominant WHO/ISUP nucleolar grade was associated with PFS (p = 0.013) and CSS (p = 0.030), the presence of non-predominant (<50%) nucleolar grade did not show association with outcome (p = 0.7). PFS and CSS showed no significant association with the presence or the amount of type 2 morphology. We compared the molecular alterations in paired type 1 and type 2 areas in a subset of 22 cases with mixed type 1 and 2 features and identified 12 recurrently mutated genes including TERT, ARID1A, KDM6A, KMT2D, NFE2L2, MET, APC, and TP53. Among 78 detected somatic mutations, 61 (78%) were shared between the paired type 1 and type 2 areas. Copy number alterations, including chromosome 7 and 17 gains, were similar between type 1 and 2 areas. These findings support that type 2 features in a PRCC with mixed histology represent either morphologic variance or clonal evolution. Our study underscores the notion that PRCC with any classic type 1 regions is best considered as type 1 PRCC and assigned the appropriate WHO/ISUP nucleolar grade. It provides additional evidence that type 2 PRCC as a separate category should be re-assessed and likely needs to be abandoned.
Collapse
|
6
|
Chan E, Stohr BA, Butler RS, Cox RM, Myles JL, Nguyen JK, Przybycin CG, Reynolds JP, Williamson SR, McKenney JK. Papillary Renal Cell Carcinoma With Microcystic Architecture Is Strongly Associated With Extrarenal Invasion and Metastatic Disease. Am J Surg Pathol 2022; 46:392-403. [PMID: 34881751 DOI: 10.1097/pas.0000000000001802] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Papillary renal cell carcinoma (PRCC) is well-recognized as a morphologically and molecularly heterogenous group of kidney tumors with variable clinical behavior. Our goal was to analyze a unique histologic pattern of PRCC we have observed in routine practice to evaluate for potential clinical significance or distinct molecular signature. We identified 42 cases of PRCC showing a morphologically distinct architecture characterized by numerous epithelial-lined cysts containing the papillary tumor (herein called "microcysts"), which are typically separated by fibrous stroma. Of the initial 42 case test set with microcystic features, 23 (55%) were stage pT3a or higher. Most tumors had strong and diffuse cytoplasmic immunoreactivity for CK7 (93%, 37/40) and AMACR (100%, 40/40). Fumarate hydratase staining was retained in all cases tested (39/39). We performed next-generation sequencing on 15 of these cases with available tissue and identified chromosomal alterations commonly reported in historically "type 1" PRCC, notably multiple chromosomal gains, particularly of chromosomes 7 and 17, and MET alterations. However, alterations in pathways associated with more aggressive behavior (including SETD2, CDKN2A, and members of the NRF pathway) were also identified in 6 of 15 cases tested (40%). Given this molecular and immunophenotypic data, we subsequently reviewed an additional group of 60 consecutive pT2b-pT3 PRCCs to allow for comparisons between cases with and without microcysts, to assess for potential associations with other recently described histologic patterns (ie, "unfavorable architecture": micropapillary, solid, and hobnail), and to assess interobserver reproducibility for diagnosing architectural patterns and grade. Of the total combined 102 PRCCs, 67 (66%) had microcystic architecture within the intrarenal component but were commonly admixed with other patterns (39% had micropapillary, 31% solid, and 31% hobnail). Twenty-seven cases (26%) had metastatic disease, and 24 of these 27 (89%) had microcystic architecture in the intrarenal tumor. Within the pT3 subset, 21 of 22 cases with metastases (95%) had extrarenal invasion as either individual microcysts in renal sinus fat or aggregates of microcysts bulging beyond the confines of the capsule. Backward elimination and stepwise regression methods to detect features significantly associated with adverse outcome identified solid architecture (hazard ratio [HR]: 6.3; confidence interval [CI]: 2.1-18.8; P=0.001), hobnail architecture (HR: 5.3; CI: 1.7-16.7; P=0.004), and microcystic architecture at the tumor-stromal interface (HR: 4.2; CI: 1.1-16.7; P=0.036) as strongest. Of architectural patterns and grade, the microcystic pattern had a substantial interobserver agreement (κ score=0.795) that was highest among the 6 observers. In summary, PRCCs with microcystic architecture represents a subset of historically "type 1" PRCC with a predilection for morphologically distinctive extrarenal involvement and metastatic disease. Microcysts co-vary with other "unfavorable" architectural patterns also associated with higher risk for aggressive disease (ie, micropapillary, hobnail, and solid), but microcysts were more common and have superior interobserver reproducibility. These findings suggest that microcystic PRCC should be recognized as a potentially aggressive histologic pattern of growth in PRCC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Papillary/diagnosis
- Carcinoma, Papillary/genetics
- Carcinoma, Papillary/metabolism
- Carcinoma, Papillary/pathology
- Carcinoma, Renal Cell/diagnosis
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cysts/diagnosis
- Cysts/genetics
- Cysts/metabolism
- Cysts/pathology
- Female
- Follow-Up Studies
- Humans
- Immunohistochemistry
- Kidney Neoplasms/diagnosis
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Male
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Metastasis
- Observer Variation
- Prognosis
- Reproducibility of Results
- Retrospective Studies
Collapse
Affiliation(s)
- Emily Chan
- Department of Pathology, University of California San Francisco (UCSF), San Francisco, CA
| | - Bradley A Stohr
- Department of Pathology, University of California San Francisco (UCSF), San Francisco, CA
| | - Robert S Butler
- Department of Quantitative Health Sciences, Cleveland Clinic
| | - Roni M Cox
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jonathan L Myles
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jane K Nguyen
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Christopher G Przybycin
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jordan P Reynolds
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Sean R Williamson
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Jesse K McKenney
- Department of Pathology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
7
|
Moch H, Ohashi R. Chromophobe renal cell carcinoma: current and controversial issues. Pathology 2020; 53:101-108. [PMID: 33183792 DOI: 10.1016/j.pathol.2020.09.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022]
Abstract
It has been 35 years since Professor Thoenes and his colleagues discovered chromophobe renal cell carcinoma (RCC). Since then, our knowledge about this tumour entity has changed and novel tumour entities have been discovered. The aim of this review is to discuss recent molecular findings and open questions in diagnosing chromophobe-like/oncocytic neoplasms. The broader differential diagnosis of chromophobe-like and oncocytoma-like neoplasms includes SDH-deficient renal cell carcinoma, fumarate hydratase (FH) deficient RCC, epitheloid angiomyolipoma ('oncocytoma like'), MiT family translocation RCC and the emerging entity of eosinophilic solid and cystic renal cell carcinoma. After separation of these tumours from chromophobe RCC, it becomes evident that chromophobe RCC are low malignant tumours with a 5-6% risk of metastasis. Recent next generation sequencing (NGS) and DNA methylation profiling studies have confirmed Thoenes' theory of a distal tubule derived origin of chromophobe RCC and renal oncocytomas. Comprehensive genomic analyses of chromophobe RCC have demonstrated a low somatic mutation rate and identified TP53 and PTEN as the most frequently mutated genes, whereas 'unclassified' RCC with oncocytic or chromophobe-like features can show somatic inactivating mutations of TSC2 or activating mutations of MTOR as the primary molecular alterations. For the future, it would be desirable to create a category of 'oncocytic/chromophobe RCC, NOS' with the potential of further molecular studies for identification of TSC1/2 mutations in these rare tumours.
Collapse
Affiliation(s)
- Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland.
| | - Riuko Ohashi
- Histopathology Core Facility, Niigata University Faculty of Medicine, Niigata, Japan
| |
Collapse
|
8
|
Delahunt B, Eble JN, Samaratunga H, Thunders M, Yaxley JW, Egevad L. Staging of renal cell carcinoma: current progress and potential advances. Pathology 2020; 53:120-128. [PMID: 33121821 DOI: 10.1016/j.pathol.2020.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/14/2022]
Abstract
Formal staging classifications for renal cell carcinoma (RCC) were first proposed in 1978 and were incorporated into the Tumour, Nodes, Metastases (TNM) system initially published by the Union Internationale Contre le Cancer (UICC) in 1978. There has been a gradual evolution of grading criteria through six separate editions of the UICC TNM Classification, with the latest edition being published in 2016. Somewhat surprisingly there were no changes to the T category criteria from the 2009 to the 2016 editions of the classification, although an erratum has subsequently been published that incorporated the minor changes included in the eighth edition of the TNM Classification published by the American Joint Committee on Cancer. Localised tumours are staged according to the size of the primary tumour, with the TNM classification recognising that these tumours may exceed 10 cm in diameter. This is unfortunate as there is good evidence to demonstrate that, for clear cell RCC, virtually all tumours >7 cm in diameter and a substantial proportion of tumours <7 cm in diameter, show extra-renal spread. Infiltration of tumour beyond the renal capsule into the peri-renal fat is also categorised as T3a, however the clinical importance of this remains unclear. The classification of microvascular invasion within the renal sinus requires clarification, as does the prognostic significance of tumour in small vessels within the kidney.
Collapse
Affiliation(s)
- Brett Delahunt
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand.
| | - John N Eble
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, USA
| | | | - Michelle Thunders
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - John W Yaxley
- Department of Medicine, University of Queensland, Wesley Urology Clinic, Royal Brisbane and Womens Hospital, Brisbane, Qld, Australia
| | - Lars Egevad
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Clark DJ, Zhang H. Proteomic approaches for characterizing renal cell carcinoma. Clin Proteomics 2020; 17:28. [PMID: 32742246 PMCID: PMC7391522 DOI: 10.1186/s12014-020-09291-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022] Open
Abstract
Renal cell carcinoma is among the top 15 most commonly diagnosed cancers worldwide, comprising multiple sub-histologies with distinct genomic, proteomic, and clinicopathological features. Proteomic methodologies enable the detection and quantitation of protein profiles associated with the disease state and have been explored to delineate the dysregulated cellular processes associated with renal cell carcinoma. In this review we highlight the reports that employed proteomic technologies to characterize tissue, blood, and urine samples obtained from renal cell carcinoma patients. We describe the proteomic approaches utilized and relate the results of studies in the larger context of renal cell carcinoma biology. Moreover, we discuss some unmet clinical needs and how emerging proteomic approaches can seek to address them. There has been significant progress to characterize the molecular features of renal cell carcinoma; however, despite the large-scale studies that have characterized the genomic and transcriptomic profiles, curative treatments are still elusive. Proteomics facilitates a direct evaluation of the functional modules that drive pathobiology, and the resulting protein profiles would have applications in diagnostics, patient stratification, and identification of novel therapeutic interventions.
Collapse
Affiliation(s)
- David J. Clark
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231 USA
| | - Hui Zhang
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231 USA
| |
Collapse
|
10
|
MacLennan GT, Cheng L. Five decades of urologic pathology: the accelerating expansion of knowledge in renal cell neoplasia. Hum Pathol 2019; 95:24-45. [PMID: 31655169 DOI: 10.1016/j.humpath.2019.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023]
Abstract
Those who are knowledgeable in cosmology inform us that the expansion of the universe is such that the velocity at which a distant galaxy is receding from the observer is continually increasing with time. We humbly paraphrase that as "The bigger the universe gets, the faster it gets bigger." This is an interesting analogy for the expansion of knowledge in the field of renal tumor pathology over the past 30 to 50 years. It is clear that a multitude of dedicated investigators have devoted incalculable amounts of time and effort to the pursuit of knowledge about renal epithelial neoplasms. As a consequence of the contributions of numerous investigators over many decades, the most recent World Health Organization classification of renal neoplasms includes about 50 well defined and distinctive renal tumors, as well as various miscellaneous and metastatic tumors. In addition, a number of emerging or provisional new entities are under active investigation and may be included in future classifications. In this review, we will focus on a number of these tumors, tracing as accurately as we can the origins of their discovery, relating relevant additions to the overall knowledge base surrounding them, and in some instances addressing changes in nomenclature.
Collapse
Affiliation(s)
- Gregory T MacLennan
- Department of Pathology and Laboratory Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH.
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
11
|
Drug resistance in papillary RCC: from putative mechanisms to clinical practicalities. Nat Rev Urol 2019; 16:655-673. [PMID: 31602010 DOI: 10.1038/s41585-019-0233-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2019] [Indexed: 11/08/2022]
Abstract
Papillary renal cell carcinoma (pRCC) is the second most common renal cell carcinoma (RCC) subtype and accounts for 10-15% of all RCCs. Despite clinical need, few pharmacogenomics studies in pRCC have been performed. Moreover, current research fails to adequately include pRCC laboratory models, such as the ACHN or Caki-2 pRCC cell lines. The molecular mechanisms involved in pRCC development and drug resistance are more diverse than in clear-cell RCC, in which inactivation of VHL occurs in the majority of tumours. Drug resistance to multiple therapies in pRCC occurs via genetic alteration (such as mutations resulting in abnormal receptor tyrosine kinase activation or RALBP1 inhibition), dysregulation of signalling pathways (such as GSK3β-EIF4EBP1, PI3K-AKT and the MAPK or interleukin signalling pathways), deregulation of cellular processes (such as resistance to apoptosis or epithelial-to-mesenchymal transition) and interactions between the cell and its environment (for example, through activation of matrix metalloproteinases). Improved understanding of resistance mechanisms will facilitate drug discovery and provide new effective therapies. Further studies on novel resistance biomarkers are needed to improve patient prognosis and stratification as well as drug development.
Collapse
|
12
|
Delahunt B, Eble JN, Egevad L, Yaxley J, Thunders M, Samaratunga H. Emerging entities of renal cell neoplasia. SURGICAL AND EXPERIMENTAL PATHOLOGY 2019. [DOI: 10.1186/s42047-019-0035-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractThe current classification of renal cell carcinoma (RCC) was formulated at the meeting of the World Health Organization Renal Tumor Panel in 2015, with the results published in the fourth edition of the World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs Bluebook in 2016. At that meeting a number of tumor types were designated as emerging or provisional entities as it was felt that they were insufficiently characterized to merit inclusion as a recognized type of RCC. One tumor type included in this designation was thyroid-like follicular RCC. Since the publication of the 2016 classification this tumor type has been further characterized and in addition to this, detailed studies on three other types of RCC (multifocal oncocytoma-like tumors associated with oncocytosis, eosinophilic solid and cystic RCC and biphasic squamoid alveolar RCC) have been published. It is now apparent that these four tumors are unique morphotypes and genotypes of RCC, and are likely to be included in the next edition of the World Health Organization classification of renal tumors. Multifocal oncocytoma-like tumors associated with oncocytosis is a benign process characterized by the presence of hundreds to thousands of oncocytic tumors in a single kidney. These tumors occur sporadically and are unrelated to the tumors of Birt-Hogg-Dubé syndrome. Eosinophilic solid and cystic RCC is characterized by a solid and cystic architecture with tumor cells consisting of bulky eosinophilic and granular cytoplasm with intracytoplasmic vacuolation. Thyroid-like follicular RCC occurs in younger patients with a female predominance. The tumor bears a striking resemblance to follicular carcinoma of the thyroid with follicles containing intraluminal proteineacous material resembling thyroglobulin. Immunostains for thyroid markers are negative. Finally, biphasic squamoid alveolar RCC consists of aggregates of large cells with pale eosinophilic cytoplasm usually arranged in a glomeruloid/alveolar pattern and surrounded by a border of basophilic cells with scanty cytoplasm. The genotype of the tumor, as well its recorded association with typical papillary RCC, has led to the suggestion that it is related to type 1 papillary RCC.
Collapse
|
13
|
Young RH, Eble JN. The history of urologic pathology: an overview. Histopathology 2019; 74:184-212. [PMID: 30565309 DOI: 10.1111/his.13753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/07/2018] [Indexed: 12/25/2022]
Abstract
This article begins with the testis and a legendary figure, Sir Astley Cooper, who wrote an early text on the organ. The early 20th century saw the first major development, the description of the seminoma by the French investigator Maurice Chevassu, but the pace of knowledge did not accelerate until after World War II with a major article from the Armed Forces Institute of Pathology (AFIP) by Nathan B. Friedman and Robert A. Moore, soon followed by the first series testis fascicle by Frank J. Dixon and Moore. Other noteworthy contributions were made by two masters of gonadal pathology, Gunnar Teilum and Robert E. Scully. In the 1970s, Niels E. Skakkebaek played a seminal role in elaborating in-situ neoplasia of the testis. The school of British testicular tumour authored, in the mid-1970s, under the editorship of Roger C. B. Pugh, one of the best texts on testicular pathology. Advances in more recent years have been largely spearheaded by Thomas M. Ulbright of the Indiana University School of Medicine. Observations on the prostate gland date back to Andreas Vesalius and William Cheselden, the latter appearing to have introduced the word for the gland. Note is made of contributions on the anatomy and histology of the gland by Oswald Lowsley, L. M. Franks, and John McNeal. Diagnosing carcinoma of the prostate was brought into the modern age in a landmark 1953 article by Robert S. Totten et al. In the 1960s, Donald F. Gleason introduced a grading system that is now in use worldwide. The topic of premalignant lesions has been well established only for approximately three decades, based initially on the work of Dr McNeal and David G. Bostwick. One of the first to write a book on the bladder was the remarkable British surgeon-pathologist Sir Henry Thompson. Workers at the AFIP, including Colonel James E. Ash and Fatallah K. Mostofi, wrote many outstanding articles on bladder pathology. The roles of other institutions, such as Johns Hopkins University, the Mayo Clinic, and St Peter's Hospital Institute of Urology, London, and those who worked there are noted. Knowledge of the pathology of the urachus dates largely back to the remarkable book on the topic in 1916 by the Hopkins investigator Thomas S. Cullen. Information on renal tumours dates largely to the work of Paul Grawitz, but awareness of the many variants of renal cell carcinoma in general was slow to evolve, and has only accelerated in recent years. The AFIP group of Dr Mostofi, ably assisted by Colonel Charles J. Davis and Isabell A. Sesterhenn, has contributed to knowledge of renal neoplasia with articles of note on oncocytoma, metanephric adenoma, and medullary carcinoma. In the mid-1980s, the German workers Wolfgang Thoenes and Stephan Störkel recognised the distinctive tumour known as chromophobe renal cell carcinoma. Work on renal tumours in the young owes much to J. Bruce Beckwith. The observational talents of numerous investigators have, in just over a century, advanced our knowledge of diseases of the urinary tract and testis remarkably.
Collapse
Affiliation(s)
- Robert H Young
- James Homer Wright Pathology Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John N Eble
- Indiana University Medical Center, Indianapolis, IN, USA
| |
Collapse
|
14
|
Abstract
Papillary renal cell carcinoma (PRCC) is the second most common type of renal carcinoma following clear cell renal cell carcinoma. Papillary renal cell carcinoma is usually divided histologically into 2 types namely, type 1 and type 2. This classification, however, is unsatisfactory as many of papillary carcinoma are unclassifiable by the existing criteria. In recent years there has been a remarkable progress in our understanding of the molecular basis of PRCC. These studies have revealed that type 2 PRCCs represent a heterogenous group which may be subdivided into additional subtypes based on the genetic and molecular make up of these tumors and reflecting different clinical course and prognosis. Some of the molecular features such a hypermethylation of CPG islands in the promotor regions of genes and over expression of the antioxidant pathways within tumor cells have been recognized as markers of poor prognosis. Targeted therapies for papillary carcinoma in the past have been unsuccessful because of lack of clear understanding of the molecular basis of these tumors. It is hoped that recent progress in our understanding of the pathogenesis of various subtypes of PRCC, effective targeted therapies will eventually emerge in due course.
Collapse
|
15
|
Carden MA, Smith S, Meany H, Yin H, Alazraki A, Rapkin LB. Platinum plus bortezomib for the treatment of pediatric renal medullary carcinoma: Two cases. Pediatr Blood Cancer 2017; 64. [PMID: 28052556 DOI: 10.1002/pbc.26402] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/01/2016] [Accepted: 11/15/2016] [Indexed: 11/10/2022]
Abstract
Renal medullary carcinoma (RMC) was first described over two decades ago as the seventh sickle nephropathy. Survival after diagnosis with metastatic disease still rarely extends beyond 1 year, with recent reports of median overall survival in patients treated with platinum therapy being just 10 months. We describe our experience using platinum-based chemotherapy plus the proteasome inhibitor bortezomib to treat metastatic RMC in two pediatric patients who had complete responses. One patient passed away 7 years after diagnosis, while another remains disease free nearly 2 years from diagnosis.
Collapse
Affiliation(s)
- Marcus A Carden
- Department of Pediatric Hematology/Oncology, Emory University School of Medicine, Children's Healthcare of Atlanta, Aflac Cancer & Blood Disorders Center, Atlanta, Georgia
| | - Stephen Smith
- Department of Pediatric Hematology/Oncology, University of Kansas Medical Center
| | - Holly Meany
- Department of Hematology/Oncology, Children's National Medical Center
| | - Hong Yin
- Department of Pathology, Children's Healthcare of Atlanta
| | - Adina Alazraki
- Department of Radiology and Imaging Sciences, Children's Healthcare of Atlanta and Emory University School of Medicine
| | - Louis B Rapkin
- Department of Pediatric Hematology/Oncology, Emory University School of Medicine, Children's Healthcare of Atlanta, Aflac Cancer & Blood Disorders Center, Atlanta, Georgia
| |
Collapse
|
16
|
Delahunt B, Samaratunga H, Kenwright DN. Histologic prognostic markers for renal cell neoplasia. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.mpdhp.2016.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
|
18
|
Bhatt JR, Finelli A. Landmarks in the diagnosis and treatment of renal cell carcinoma. Nat Rev Urol 2014; 11:517-25. [PMID: 25112856 DOI: 10.1038/nrurol.2014.194] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The most common renal cancer is renal cell carcinoma (RCC), which arises from the renal parenchyma. The global incidence of RCC has increased over the past two decades by 2% per year. RCC is the most lethal of the common urological cancers: despite diagnostic advances, 20-30% of patients present with metastatic disease. A clearer understanding of the genetic basis of RCC has led to immune-based and targeted treatments for this chemoresistant cancer. Despite promising results in advanced disease, overall response rates and durable complete responses are rare. Surgery remains the main treatment modality, especially for organ-confined disease, with a selective role in advanced and metastatic disease. Smaller tumours are increasingly managed with biopsy, minimally invasive interventions and surveillance. The future promises multimodal, integrated and personalized care, with further understanding of the disease leading to new treatment options.
Collapse
Affiliation(s)
- Jaimin R Bhatt
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| | - Antonio Finelli
- Princess Margaret Cancer Centre, University of Toronto, Division of Urology, 610 University Avenue 3-130, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
19
|
Kurosch M, Reiter M, Haferkamp A. Epidemiologie, Diagnostik und chirurgische Therapie des Nierenzellkarzinoms. DER ONKOLOGE 2014. [DOI: 10.1007/s00761-014-2750-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Goyal R, Gersbach E, Yang XJ, Rohan SM. Differential diagnosis of renal tumors with clear cytoplasm: clinical relevance of renal tumor subclassification in the era of targeted therapies and personalized medicine. Arch Pathol Lab Med 2013; 137:467-80. [PMID: 23544936 DOI: 10.5858/arpa.2012-0085-ra] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT The World Health Organization classification of renal tumors synthesizes morphologic, immunohistochemical, and molecular findings to define more than 40 tumor types. Of these, clear cell (conventional) renal cell carcinoma is the most common malignant tumor in adults and-with the exception of some rare tumors-the most deadly. The diagnosis of clear cell renal cell carcinoma on morphologic grounds alone is generally straightforward, but challenging cases are not infrequent. A misdiagnosis of clear cell renal cell carcinoma has clinical consequences, particularly in the current era of targeted therapies. OBJECTIVE To highlight morphologic mimics of clear cell renal cell carcinoma and provide strategies to help differentiate clear cell renal cell carcinoma from other renal tumors and lesions. The role of the pathologist in guiding treatment for renal malignancies will be emphasized to stress the importance of proper tumor classification in patient management. DATA SOURCES Published literature and personal experience. CONCLUSIONS In challenging cases, submission of additional tissue is often an inexpensive and effective way to facilitate a correct diagnosis. If immunohistochemical stains are to be used, it is best to use a panel of markers, as no one marker is specific for a given renal tumor subtype. Selection of limited markers, based on a specific differential diagnosis, can be as useful as a large panel in reaching a definitive diagnosis. For renal tumors, both the presence and absence of immunoreactivity and the pattern of labeling (membranous, cytoplasmic, diffuse, focal) are important when interpreting the results of immunohistochemical stains.
Collapse
Affiliation(s)
- Rajen Goyal
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | | | |
Collapse
|
21
|
You D, Shim M, Jeong IG, Song C, Kim JK, Ro JY, Hong JH, Ahn H, Kim CS. Multilocular cystic renal cell carcinoma: clinicopathological features and preoperative prediction using multiphase computed tomography. BJU Int 2011; 108:1444-9. [PMID: 21722289 DOI: 10.1111/j.1464-410x.2011.10247.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE • To analyse the clinicopathological and radiological features of multilocular cystic renal cell carcinoma (MCRCC) and to determine the preoperative factors differentiating MCRCC from other cystic RCC (CRCC). PATIENTS AND METHODS • The medical records of 53 patients with complex cystic renal masses evaluated by multiphase computed tomography (CT), surgically removed and confirmed as sporadic RCC were reviewed. • Of these 53 patients, 23 were classified as having MCRCC and 30 as other CRCCs, defined as RCCs with extensive cystic change or cystic necrosis. • Another 22 patients were treated for complex cystic renal masses presumed to be RCC and diagnosed as having benign cyst. RESULTS • Benign cysts and MCRCCs were significantly more likely to be of Bosniak classification III than other CRCCs (77% vs 61% vs 27%, P= 0.001). • The mean Hounsfield unit (HU) during the corticomedullary phase (CMP) was significantly higher in other CRCCs, with HU ≥38 having 83% sensitivity and 80% specificity for predicting other CRCCs. • In a multiple regression model, Bosniak classification and mean HU during CMP were independent factors predictive of other CRCCs. • In the 41 patients with masses >4 cm in diameter, the combination of Bosniak classification IV and HU ≥38 during CMP showed 63% sensitivity, 96% specificity, 91% positive predictive value and 80% negative predictive value, yielding 2% false-positive and 15% false-negative rates. CONCLUSIONS • The mean HU during CMP and Bosniak classification can differentiate MCRCC from other CRCCs. • This could help in selecting an appropriate surgical method, such as nephron-sparing surgery, for complex cystic renal masses >4 cm.
Collapse
Affiliation(s)
- Dalsan You
- Department of Urology, Radiology Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Renal cell carcinoma with novel VCL-ALK fusion: new representative of ALK-associated tumor spectrum. Mod Pathol 2011; 24:430-42. [PMID: 21076462 DOI: 10.1038/modpathol.2010.213] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Renal cell carcinoma represents a model for contemporary classification of solid tumors; however, unusual and unclassifiable cases exist and are not rare in children and young adults. The anaplastic lymphoma kinase (ALK) gene has recently been implicated in subsets of pulmonary, esophageal, breast, and colon cancers. These findings strengthen the importance of molecular classification of carcinomas across different organ sites, especially considering the evolving targeted anticancer therapies with ALK inhibitors. In the current study of six pediatric renal cell carcinomas, two cases exhibited structural karyotypic abnormalities involving the ALK locus on chromosomal band 2p23. Fluorescence in situ hybridization (FISH) studies were positive for an ALK rearrangement in one case, and subsequent 5' rapid amplification of cDNA ends analysis of this tumor revealed that the 3' portion of the ALK transcript encoding for the kinase domain was fused in frame to the 5' portion of vinculin (VCL, NM_003373). The new fusion gene is predicted to have an open reading frame of 4122 bp encoding for a 1374-aa oncoprotein; its expression was shown by immunoblotting with anti-VCL and anti-ALK antibodies in tumor tissue lysates. Immunohistochemistry with the same antibodies demonstrated cytoplasmic and subplasmalemmal localization of the oncoprotein determined by its N-terminal VCL portion. FISH with a custom-designed VCL-ALK dual-fusion probe set confirmed the presence of the fusion in neoplastic cells and demonstrated the potential clinical utility of this approach for detecting VCL-ALK in routinely processed tissue. The five remaining pediatric renal cell carcinomas did not show ALK rearrangement by FISH or ALK expression by immunohistochemistry. The data identify the kidney as a new organ site for ALK-associated carcinomas and VCL as a novel ALK fusion partner. The results should prompt further studies to advance the molecular classification of renal cell carcinoma and help to select patients who would benefit from appropriate targeted therapies.
Collapse
|
23
|
Affiliation(s)
- A Haferkamp
- Klinik fur Urologie, Universitatsklinikum Im Neuenheimer Feld 110, 69120 Heidelberg.
| | | | | | | | | |
Collapse
|
24
|
Abstract
Major consensus conferences held over a decade ago laid the foundations for the current (2004) WHO classification of renal carcinoma. Clear cell, papillary and chromophobe carcinomas account for 85-90% carcinomas seen in routine practice. The remaining 10-15% of carcinomas consist of rare sporadic and hereditary tumors, some of which had been long recognized, but many of which only emerged as distinct entities in the decade leading up to the WHO publication. Collecting-duct carcinoma is a rare, often lethal form of carcinoma. Medullary carcinoma associated with sickle cell trait, has emerged as a distinctive tumor showing some overlapping features with upper tract urothelial carcinoma. Mucinous tubular and spindle-cell carcinoma and tubulocystic carcinoma were earlier considered as patterns of low-grade collecting-duct carcinoma, but are now recognized as separate tumor entities. Carcinomas associated with somatic translocations of TFE3 and TFEB comprise a significant proportion of pediatric renal carcinomas. Oncocytoid renal carcinomas in neuroblastoma survivors was recognized as a unique tumor category in the WHO classification. Renal carcinoma associated with end-stage renal disease is now recognized as having distinct morphological patterns and behavior. In addition there is a group of rare recently described carcinomas, including clear cell papillary carcinoma, oncocytic papillary renal cell carcinoma, follicular renal carcinoma and leiomyomatous renal cell carcinoma. It behooves the surgical pathologist to not only be capable of diagnosing the common forms of renal cancer, but also to be aware of the rare types of renal carcinoma, many of which have emerged in recent years.
Collapse
Affiliation(s)
- John R Srigley
- Department of Pathology and Molecular Medicine, McMaster University, c/o The Credit Valley Hospital, 2200 Eglinton Avenue West, Mississauga, ON L5M2N1, Canada.
| | | |
Collapse
|
25
|
Current insights in renal cell cancer pathology. Urol Oncol 2008; 26:225-38. [DOI: 10.1016/j.urolonc.2007.05.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2007] [Revised: 05/15/2007] [Accepted: 05/15/2007] [Indexed: 01/09/2023]
|
26
|
Montironi R, Scarpelli M, Martignoni G, Cheng L, Lopez-Beltran A. Splitting and Lumping Adult Renal Epithelial Tumours: Is That What the Urologists Want? Eur Urol 2008; 53:673-5; discussion 676-80. [DOI: 10.1016/j.eururo.2007.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 11/05/2007] [Indexed: 11/26/2022]
|
27
|
Allory Y, Bazille C, Vieillefond A, Molinié V, Cochand-Priollet B, Cussenot O, Callard P, Sibony M. Profiling and classification tree applied to renal epithelial tumours. Histopathology 2007; 52:158-66. [PMID: 18036175 DOI: 10.1111/j.1365-2559.2007.02900.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIMS Selection of the relevant combination from a growing list of candidate immunohistochemical biomarkers constitutes a real challenge. The aim was to establish the minimal subset of antibodies to achieve classification on the basis of 12 antibodies and 309 renal tumours. METHODS AND RESULTS Seventy-nine clear cell (CC), 88 papillary (PAP) and 50 chromophobe (CHRO) renal cell carcinomas, and 92 oncocytomas (ONCO) were immunostained for renal cell carcinoma antigen, vimentin, cytokeratin (CK) AE1-AE3, CK7, CD10, epithelial membrane antigen, alpha-methylacyl-CoA racemase (AMACR), c-kit, E-cadherin, Bcl-1, aquaporin 1 and mucin-1 and analysed by tissue microarrays. First, unsupervised hierarchical clustering performed with immunohistochemical profiles identified four main clusters-cluster 1 (CC 67%), 2 (PAP 98%), 3 (CHRO 67%) and 4 (ONCO 100%)-demonstrating the intrinsic classifying potential of immunohistochemistry. A series of classification trees was then automatically generated using Classification And Regression Tree software. The most powerful of these classification trees sequentially used AMACR, CK7 and CD10 (with 86% CC, 87% PAP, 79% CHRO and 78% ONCO correctly classified in a leave-one-out cross-validation test). The classifier was also helpful in 22/30 additional cases with equivocal features. CONCLUSION The classification tree method using immunohistochemical profiles can be applied successfully to construct a renal tumour classifier.
Collapse
Affiliation(s)
- Y Allory
- AP-HP, Hôpital Henri Mondor, Département de Pathologie, INSERM, IMRB U841, Créteil, France.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Delahunt B, Bethwaite PB, Nacey JN. Outcome prediction for renal cell carcinoma: evaluation of prognostic factors for tumours divided according to histological subtype. Pathology 2007; 39:459-65. [PMID: 17886093 DOI: 10.1080/00313020701570061] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A wide variety of parameters have been investigated for their prognostic significance in mixed series of renal cell carcinoma (RCC). The classification of RCC into separate types with differing morphology, genotype and probable clinical outcome has led to a re-evaluation of many prognostic parameters with studies confined to a single RCC morphotype. Tumour stage remains the most important predictor of RCC outcome and recent investigations have focused upon tumour diameter and the prognostic significance of stromal, vascular and lymphatic invasion within the renal sinus. In large tumour series, morphotype has been correlated with patient survival, with clear cell RCC being associated with a less favourable outcome than chromophobe RCC and to a lesser extent papillary RCC, for organ confined tumours. The prognostic significance of nuclear grading remains controversial. Fuhrman grading has been shown to have prognostic utility for clear cell RCC in some series. Recent studies have shown that for papillary RCC, grading should be based upon nucleolar size and that Fuhrman grading is inappropriate for chromophobe RCC. Proliferative indices based upon a variety of markers have been correlated with outcome for clear cell RCC (Ki-67, AgNORs, p21(waf1/cip1) and p27(Kip1)) and papillary RCC (Ki-67, AgNORs), although in some series prognostic significance was lost on multivariate analysis. The presence of tumour necrosis has been shown to predict survival for clear cell and chromophobe RCC, and in clear cell RCC quantification of tumour vascular density has been correlated with outcome. Several molecular markers have been investigated for prognostic significance, mostly in clear cell RCC. Although some of these markers have been shown to be significantly associated with survival, these findings remain to be confirmed in large scale follow-up studies.
Collapse
Affiliation(s)
- Brett Delahunt
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, Wellington South, New Zealand.
| | | | | |
Collapse
|
29
|
Laber DA. Risk factors, classification, and staging of renal cell cancer. Med Oncol 2007; 23:443-54. [PMID: 17303902 DOI: 10.1385/mo:23:4:443] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2006] [Revised: 11/30/1999] [Accepted: 05/31/2006] [Indexed: 01/04/2023]
Abstract
Knowledge about renal cell carcinoma (RCC) has increased exponentially over the last decades. A clear understanding of RCC is of utmost importance to prevent the disease and improve the outcomes. Large epidemiologic studies have identified cigarette smoking, chemical agents, obesity, hypertension, and end-stage renal disease as risk factors associated with RCC. Identification and confirmation of risk factors may be projected into preventive strategies. Genetic studies of inherited disorders associated with an enhanced risk of RCC have elucidated many important targets for anticancer therapy. The World Health Organization (WHO) has recently developed a new histologic classification of renal cell tumors that has demonstrated prognostic utility. A refined clinical staging system is improving our ability to prognosticate the outcome of RCC patients. This article provides a practical yet comprehensive review of the risk factors, classification, and staging of RCC focusing on recent updates.
Collapse
Affiliation(s)
- Damian A Laber
- Division of Hematology and Medical Oncology, University of Louisville, J.G. Brown Cancer Center, Louisville, KY 40202, USA.
| |
Collapse
|