Next Article in Journal
What Is a Sarcoma ‘Specialist Center’? Multidisciplinary Research Finds an Answer
Previous Article in Journal
Characterization of FOLH1 Expression in Renal Cell Carcinoma
Previous Article in Special Issue
Urine-Based Biomarker Test Uromonitor® in the Detection and Disease Monitoring of Non-Muscle-Invasive Bladder Cancer—A Systematic Review and Meta-Analysis of Diagnostic Test Performance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnostic Biomarkers in Renal Cell Tumors According to the Latest WHO Classification: A Focus on Selected New Entities

by
Francesca Sanguedolce
1,
Roberta Mazzucchelli
2,
Ugo Giovanni Falagario
3,4,
Angelo Cormio
5,*,
Magda Zanelli
6,
Andrea Palicelli
6,
Maurizio Zizzo
7,
Albino Eccher
8,
Matteo Brunelli
9,
Andrea Benedetto Galosi
5,
Giuseppe Carrieri
3 and
Luigi Cormio
3,10
1
Pathology Unit, Policlinico Foggia, University of Foggia, 71122 Foggia, Italy
2
Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health, United Hospitals, Università Politecnica delle Marche, 60126 Ancona, Italy
3
Department of Urology and Renal Transplantation, Policlinico Foggia, University of Foggia, 71122 Foggia, Italy
4
Department of Molecular Medicine and Surgery, Karolinska Institutet, 17177 Stockholm, Sweden
5
Department of Urology, Azienda Ospedaliero-Universitaria Ospedali Riuniti Di Ancona, Università Politecnica Delle Marche, Via Conca 71, 60126 Ancona, Italy
6
Pathology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
7
Surgical Oncology Unit, Azienda USL-IRCCS di Reggio Emilia, 42123 Reggio Emilia, Italy
8
Department of Pathology and Diagnostics, University and Hospital Trust of Verona, 37126 Verona, Italy
9
Department of Pathology and Diagnostics and Public Health, Section of Pathology, University Hospital of Verona, 37126 Verona, Italy
10
Department of Urology, Bonomo Teaching Hospital, 76123 Andria, Italy
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(10), 1856; https://doi.org/10.3390/cancers16101856
Submission received: 14 April 2024 / Revised: 2 May 2024 / Accepted: 9 May 2024 / Published: 13 May 2024
(This article belongs to the Special Issue New Insights into Urologic Oncology)

Abstract

:

Simple Summary

The latest World Health Organization classification for kidney tumors, released in 2022, brings important changes, introducing new types of tumors and refining the names of certain diseases. This classification highlights the need for both traditional and modern methods in diagnosing these tumors, including molecular and genetic analysis alongside conventional microscopic examination. Our article focuses on the role of specific markers identified through immunohistochemistry, a technique used to study proteins in tissues, in diagnosing these new tumors. We critically discuss the features of these tumors, such as the clear cell capillary renal cell tumor, eosinophilic solid and cystic renal cell carcinoma, and other oncocytic tumors. By understanding these markers and their significance, pathologists can better diagnose these tumors, leading to improved treatment strategies and outcomes for patients. Our review provides valuable insights into these novel tumors, offering guidance for healthcare professionals in their daily practice, ultimately benefiting society by enhancing diagnostic accuracy and patient care.

Abstract

The fifth edition of the World Health Organization (WHO) classification for urogenital tumors, released in 2022, introduces some novelties in the chapter on renal epithelial tumors compared to the previous 2016 classification. Significant changes include the recognition of new disease entities and adjustments in the nomenclature for certain pathologies. Notably, each tumor entity now includes minimum essential and desirable criteria for reliable diagnosis. This classification highlights the importance of biological and molecular characterization alongside traditional cytological and architectural features. In this view, immunophenotyping through immunohistochemistry (IHC) plays a crucial role in bridging morphology and genetics. This article aims to present and discuss the role of key immunohistochemical markers that support the diagnosis of new entities recognized in the WHO classification, focusing on critical topics associated with single markers, in the context of specific tumors, such as the clear cell capillary renal cell tumor (CCPRCT), eosinophilic solid and cystic renal cell carcinoma (ESC-RCC), and so-called “other oncocytic tumors”, namely the eosinophilic vacuolated tumor (EVT) and low-grade oncocytic tumor (LOT). Their distinctive characteristics and immunophenotypic profiles, along with insights regarding diagnostic challenges and the differential diagnosis of these tumors, are provided. This state-of-the-art review offers valuable insights in biomarkers associated with novel renal tumors, as well as a tool to implement diagnostic strategies in routine practice.

1. Introduction

The fifth edition of the World Health Organization (WHO) classification for urogenital tumors, commonly referred to as the “Blue Book” and released in 2022, provides peculiar revisions in the field of renal cell carcinoma (RCC) compared to the previous 2016 classification [1].
The most significant changes include the introduction of new disease entities, as well as changes in the nomenclature for some pathologies (see Table 1). Another distinctive feature of the fifth edition of the WHO classification is the introduction, for each tumor entity, of minimum essential and desirable criteria that enable a reliable diagnosis of them [2].
Overall, this new classification provides a more precise distinction of the various subgroups of tumors and emphasizes the importance of their biological and molecular characterization alongside traditional cytological and architectural features. In this setting, immunophenotyping (i.e., the identification of tissue biomarkers through immunohistochemistry (IHC)) plays an even greater role than in the past, as it represents a bridge between morphology and genetics through the determination of diagnostic protein biomarkers identifiable in tissue sections [3]. Since there is no specific immunophenotypic biomarker for a particular histotype of renal tumor, it is essential to apply a panel of markers that, based on morphological characteristics, allow us to favor or rule out the diagnostic hypotheses formulated on a case-by-case basis.
The aim of this paper is to present and discuss the role of the main immunohistochemical markers that often provide substantial support to the diagnosis of the new entities recognized in the current WHO classification, with a specific focus on critically analyzing the advantages and disadvantages associated with the use of individual markers.

2. Clear Cell Papillary Renal Cell Tumor (CCPRCT)

The CCPRCT was initially identified as a tumor associated with the end-stage renal disease setting and only later found in sporadic cases [4,5]. The initial designation of carcinoma was due to its significant morphological similarities to clear cell renal cell carcinoma (CCRCC), and for this reason it may be diagnostically challenging in routine practice [6]. Nevertheless, the identification of the lack of von Hippel Lindau (VHL) gene alterations, chromosome 3p35 loss, and trisomy 7/17, along with follow-up data indicating an indolent clinical behavior, with no evidence of any recurrence or metastasis so far, supported the name change from “carcinoma” to “tumor” in the new classification [7,8]. Xu et al. highlighted its genomic stability and distinct metabolic phenotype, with severe depletion of mitochondrial DNA resulting in oxidative stress [9]; such findings further highlight the molecular-level distinction between the CCPRCT and more common and morphologically similar renal epithelial malignancies, CCRCC, and papillary renal cell carcinoma (PRCC).
Since the correct diagnosis of the CCPRCT results in complete tumor excision being curative, with no need of any further treatment, its accurate diagnosis is of pivotal importance.
At gross examination, these are small (pT1), cortical, well-circumscribed lesions, which can be multiple in as many as one-fifth of all patients [8]. Microscopically, low-grade (1–2 WHO/ISUP) clear cells with suprabasal nuclei in a linear array above the basement membrane are arranged in a variable mixture of cystic, branched, acinar and tubular, solid, papillary formations. Lack of tumor necrosis or perineural and lymphovascular invasion is a constant finding.

2.1. Immunophenotype

Positivity for CK7 (diffuse staining), CAIX (“cup-shaped” expression, with staining of the basolateral but not the luminal portion of tumor cells), GATA3, PAX2, PAX8, 34βE12, AE1/AE3, or CAM5.2.
Negativity for AMACR, CD10, RCCm, or cathepsin K [4,5,10,11] (Figure 1).

2.2. Critical Topics and Insights

The assessment of CAIX, CK7, and CD10 immunostaining is regarded as a desirable diagnostic criterion in these tumors, and therefore it is advisable to apply a minimal panel consisting of these three biomarkers in routine practice, to be expanded both in challenging cases [8] and in those rare instances where combined features of CCRCC and the CCPRCT may occur [12]. In the latter case, it has been recommended to label the tumor as CCRCC if the immunophenotype does not perfectly match. The possible presence of “mixed” tumors might affect a straightforward diagnosis of the CCPRCT, especially in the biopsy setting [12]. Notably, according to the results from a recent ISUP consensus conference, IHC was used as a confirmatory tool even in morphologically typical cases by 77.4% of respondents [12], with molecular studies being advocated as second-line diagnostic tests, to be applied only in equivocal cases.
The “cup-shaped” CAIX staining is considered typical of the CCPRCT, unlike the complete membranous pattern of labeling in CCRCC. Nevertheless, such basolateral CAIX expression has been described, though occasionally, in CCRCC as well, especially in pseudopapillary areas, which are morphological mimickers of the CCPRCT [13].
Recently, CD10 and AMACR positivity has been described in as many as 53% and 35% of CCPRCT cases, respectively, in a study on cytology specimens [14]. Furthermore, AMACR has been recently reported as focally positive in cystic spaces or papillary fronds of the CCPRCT [13].
The frequent expression of 34βE12 and GATA3 suggests that these tumors, unlike CCRCC, may derive from the distal rather than proximal nephron, despite their clear cell morphology [15]. GATA3 staining raises the diagnostic possibility of a urothelial carcinoma (UC), especially upon renal biopsy; moreover, UCs of the upper urinary tract may be PAX8-positive in approximately 20% of cases [16].
The main differential diagnosis of the CCPRCT is with other renal tumors which may exhibit low-grade, clear cell, and/or papillary morphology, such as CCRCC and PRCC (see Table 2). In a broader context, the very rare ELOC-mutated RCC has overlapping morphological features with the CCPRCT, namely low stage and grade, the presence of branching tubules and papillae, and clear cytoplasm. Like the CCPRCT, ELOC-mutated RCC is CK7/CAIX/CK34βE12-positive, and the distinction between these two entities relies on a lack of prominent subnuclear vacuolization and the presence of CD10 staining in the latter [17].

3. Eosinophilic Solid and Cystic Renal Cell Carcinoma (ESC-RCC)

In the current WHO classification, eosinophilic solid and cystic renal cell carcinoma (ESC-RCC) has been introduced as a new entity, under the category “Other renal tumours”. These cortical tumors are mainly pT1, well-circumscribed, sporadic, and solitary lesions, often incidentally diagnosed [18,19]. Microscopically, they show a solid and cystic appearance, and tumor cells in solid areas have abundant eosinophilic cytoplasm, whereas hobnail cells line the cystic spaces. Under electron microscopy, characteristic aggregates of rough endoplasmic reticulum and granular material are seen in tumor cells, appearing as basophilic to dark, coarse, intracytoplasmic granules (stippling) under light microscopy. Other morphological findings are psammoma bodies and intracytoplasmic vacuoles [20].
Typically, biallelic losses or mutations in the TSC1/TSC2 genes are found in ESC-RCC, resulting in activation of mTOR complex 1; only few cases have been described in patients with tuberous sclerosis complex (TSC) [21,22]. Due to its peculiar molecular setting, a correct diagnosis of ESC-RCC is of pivotal importance for patient selection in targeted therapy with mTOR inhibitors [23].
ESC-RCC has a benign clinical behavior in most cases, with few metastatic cases being reported so far [23,24]; such unfavorable behavior was associated with larger size and morphological aggressive features, namely necrosis and hemorrhage [23,24,25].

3.1. Immunophenotype

Positivity for cathepsin k (usually patchy, focal to diffuse), CK20 (patchy, focal to diffuse, may be negative in up to 20% of cases), PAX8, AE1/AE3 (rarely focal to negative), CK8/18, vimentin, or melan-A (occasionally).
Negativity for CK7 (rarely focal expression), CD117(KIT), CAIX, or HMB45 [19,23,26,27].

3.2. Critical Topics and Insights

While the typical constellation of morphological features is an essential diagnostic criterion of ESC-RCC, CK20 immunoreactivity along with mutations in TSC1 or TSC2 genes are listed among desirable criteria [18].
The combined CK20pos/CK7neg staining in ESC-RCC is unique among RCC subtypes [26]. Furthermore, even the uncommon CK20-negative cases are not simultaneously CK7-positive, allowing for a distinction from more frequent oncocytic renal tumors, such as chromophobe RCC (ChrRCC) [28].
Li et al. reported that approximately 30% of cases from a series of 33 patients aged 35 years or younger diagnosed with “unclassified” RCCs with a predominantly eosinophilic cytoplasm were reclassified as ESC-RCCs, thus prompting the need to perform CK20 IHC in young patients with oncocytic tumors [29]. Notably, Akgul et al. recently investigated the immunophenotypes of 300 cases of RCC that were unclassified, finding CK20 staining in 5 cases, which did not show morphologic features of ESC-RCC, including cystic growth or coarse intracytoplasmic granules [30].
All in all, the peculiar combination of positivity for CK20 and cathepsin K as well as the lack of staining for CK7 and CD117 allows us to reliably diagnose ESC-RCC in the proper clinical and morphological setting. Since it is a still poorly known histotype, especially in a sporadic setting, some of its biological and clinical features are yet to be defined, which will likely occur only over time.

4. Oncocytic Tumors

The current classification of renal epithelial tumors introduces a category of “Oncocytic and chromophobe renal cell tumors”, which encompasses two well-known diagnostic entities, namely renal oncocytoma (RO) and ChrRCC, along with two emerging entities (collectively referred to as “Other oncocytic tumors of the kidney”), namely the low-grade oncocytic tumor (LOT) and the eosinophilic vacuolated tumor (EVT). The latter two tumors exhibit several overlapping features: the presence of eosinophilic cells, indolent behavior, and distinct molecular alterations, such as inactivating somatic mutations of the TSC2 gene or activating mutations of mTOR, as seen in ESC-RCC [31,32]. The presence of an overlapping genetic profile in these three tumors might lead to the proposal of grouping them separately into the same category, but the significant morphological and immunohistochemical differences among them, as well as the fact that such molecular alterations have been identified in other renal tumors, do not support this hypothesis [15].
As for now, the differential diagnosis between these two tumors does not have significant implications in clinical practice [33]. Nevertheless, since they are emerging entities, which, most probably, have likely been misdiagnosed in the past as other histotypes of oncocytic tumors or as unclassified RCCs, we cannot reliably determine their incidence or biological behavior, at least until these tumors have fully entered routine diagnostics and can be the subject of further clinical and molecular studies. All in all, the aim of this new classification is to keep these two tumors in a separate subgroup from tumors labeled as “Renal cell carcinoma NOS”, in order to reduce the latter wastebasket category for use with highly aggressive tumors only [1].
From the practicing pathologist’s point of view, the distinction among entities in the spectrum of oncocytic tumors may be challenging when handling situations with limited tissue in needle biopsy specimens. In a recent state-of-the-art review of the LOT [34], a protocol was suggested in this setting, including meticulous evaluation of morphology alongside the immunohistochemical profile (see Table 3), in order to achieve the most accurate diagnosis possible. However, in cases where this approach may not be feasible, the pathology report will include a descriptive diagnosis like “oncocytic tumor, not further specified”, accompanied by a comment detailing the list of potential differential diagnoses and/or preferences [34].

4.1. Eosinophilic Vacuolated Tumor (EVT)

The eosinophilic vacuolated tumor (EVT), previously known as the high-grade oncocytic tumor [35] or RCC with eosinophilic vacuolated cytoplasm [36], is an uncommon entity, with more than 50 cases reported to date, all exhibiting a benign behavior, with no evidence of recurrences or metastases [19,28,32,35,37,38].
The EVT is a solitary and sporadic tumor, only rarely presenting in patients with TSC, and it is not larger than 7 cm (mean tumor size between 3 and 4 cm) [19,28,32,35,36,39,40]. Under light microscopy, tumor cells have an abundant eosinophilic cytoplasm with prominent intracytoplasmic vacuoles, and round to oval nuclei with striking, sometimes large nucleoli, falling into the category of nuclear grade WHO/ISUP 3, and peripheral thick-walled vessels are a common finding [35,36,38]. The oncocytic appearance of the cytoplasm is due to an increased number of intracytoplasmic mitochondria, as with an oncocytoma and the LOT, as highlighted by ultrastructural analysis [28].
Mutations in the TSC/MTOR pathway leading to mTORC1 activation have been consistently reported in the EVT, with a recent study describing non-overlapping mutations in MTOR, TSC2, and TSC1 in all cases, alongside with low mutational rates [38]. Limited data exist on the occurrence of losses of chromosomes 1 and 19p in this tumor [32].

4.1.1. Immunophenotype

Positivity for CD117 (KIT), CD10, cathepsin K (sometimes focally), CKs, PAX8, FH, or SDHB
Negativity for CK7 (can be positive only in rare, scattered cells), CK20, HMB45, Melan A, vimentin (can be positive only in rare, scattered cells), or TFE3 [19,35,38].

4.1.2. Critical Topics and Insights

EVT cells are mostly CD117-positive, and they are negative or positive only in rare cells for CK7 and, likewise, RO. However, the distinction between these two tumors is supported by (1) the different expression of cathepsin K (which is often positive in the EVT, while usually negative in RO) and CD10 (present in the EVT, absent in RO) and (2) the presence of widespread, high-grade morphological features, which rules out the diagnosis of RO [15,28,41].
The differential diagnosis between the EVT and ChrRCC is easier to make, in that it relies on all the previous markers plus CK7, which is typically, diffusely, and strongly expressed in ChrRCC; furthermore, the latter tumor lacks prominent cytoplasmic vacuoles and striking nucleoli, whereas the EVT does not feature typical ChrRCC raisinoid nuclei with perinuclear halos [28,31]. Interestingly, as many as 2% of eosinophilic tumors from the series of 300 unclassified RCC reviewed by Akgul et al. showed both morphological (high nuclear grade, intracytoplasmic vacuoles) and immunophenotypical (diffuse cathepsin K expression) features consistent with those of the EVT [30].
CD117 expression may be variable in the EVT, though present in the majority of cases [19]. The uncommon CD117-negative cases enter a differential diagnosis of angiomyolipomas (AMLs), which are CK7-negative and cathepsin K-positive, prompting the need for adding further markers (cytokeratins, PAX8, or both), especially in a biopsy setting.

4.2. Low-Grade Oncocytic Tumor (LOT)

The low-grade oncocytic tumor (LOT) is typically a single, sporadic tumor with intermediate features between RO and ChrRCC [15,19,32,41,42]. It can be multifocal in the end-stage disease and TSC setting [39,43,44,45]. According to reports so far, its clinical behavior is indolent [42,43,44,45,46]. At a molecular level, from the limited data available to date, the LOT seems to share some features with RO such as the loss of 1p36 and a diploid pattern. The common involvement of the mTOR pathway genes has been demonstrated in this tumor, further supported by the expression of the mTOR pathway activation markers p-S6 and p-4EBP1 [37,44,45,47]; conversely, these tumors lack complete chromosomal gains or losses, as well as CCND1 rearrangements [43].
Grossly, the LOT is a smaller, solid tumor with a mahogany-brown to tan cut surface (similar to that of RO). Under light microscopy, the most common growth patterns are solid and compact-nested, focal tubular, tubulo-reticular, or trabecular [41,42,43,46]. The tumor cells are eosinophilic due to the presence of abundant, closely packed cytoplasmic mitochondria, similar to those of an oncocytoma and the EVT, which can be identified under electron microscopy [28,40]. Nuclei are bland, round to oval, sometimes showing perinuclear clearing (halos), with Muller–Mowry colloidal iron staining being either negative or only luminal-positive. Oftentimes, sharply delineated, edematous, and hemorrhagic areas with loose cell growth (“boats in a bay”) can be seen [19,42]. Recently, Gupta et al. raised the issue of a tumor with morphologic overlap between the LOT and eosinophilic variant of ChrRCC, including prominent raisinoid nuclei and occasional binucleation, while both its immunoprofile and clinical behavior were consistent with those of the former [48]. The LOT lacks aggressive morphological features, such as necrosis, severe nuclear atypia, and increased mitotic activity.

4.2.1. Immunophenotype

Positivity for CK7 (strong and diffuse), GATA3, p-S6 and p-4EBP1 (at least focally), AE1/AE3, PAX8, E-cadherin, BerEP4, MOC31, FH, or SDHB.
Negativity for CD117, CAIX, CK20, CK5/6, p63, CD15, HMB45, melan-A, vimentin (may be positive), CD10 (may be focally positive), AMACR (may be focally positive), or cathepsin K [32,42,43,45,47] (Figure 2).

4.2.2. Critical Topics and Insights

While some authors suggested that typical morphological features alone allow for the identification of the LOT under light microscopy, others advocate for the use of a 2-marker antibody panel consisting of CD117 and CK7 to reliably differentiate this tumor from other oncocytic tumors, mainly RO and ChrRCC [49]. Nevertheless, there has been a recent suggestion that a distinct entity, referred to as “oncocytic renal neoplasm with diffuse keratin 7 immunohistochemistry”, shows the same immunoprofile along with RO-like morphology, and its connection with the LOT is still debatable [50].
The immunophenotype of the LOT has been investigated in two homogeneous series of 28 and 23 tumors, respectively, by Trpkov et al. [42] and Akgul et al. [46]. All reported cases were CK7-positive, whereas CD117 staining was found in 7 out of 28 tumors in one series only [42]. Furthermore, vimentin expression was diffusely present in most of the positive cases, accounting for 35% of the cases described by Akgul et al. [46]. Notably, approximately 73% of ROs from a large series were reported as vimentin-positive, with stained cells mostly at the edge of a central scar or scattered throughout [51], thus supporting its ineffectiveness as a marker in the differential diagnosis of oncocytic tumors.
GATA3 has been reported as a consistently positive marker in the LOT [47,52]. GATA3 staining, though focal and variable, has been reported in ChrRCC, as well as in a small subset of ROs from a pan-tumor immunohistochemical study [53]; nevertheless, since this study dates back to 10 years ago, it has been suggested that at least some of the cases diagnosed as ChrRCC and RO at that time, if reviewed today, would be reclassified into other diagnostic categories within the spectrum of oncocytic tumors, including the LOT [34].

5. Immunohistochemical Biomarkers in Selected Renal Tumors: A Primer

The main markers discussed in this review are summarized in Table 4.
Carbonic anhydrase IX (CAIX) is a transmembrane isozyme belonging to the carbonic anhydrase family involved in the regulation of intra- and extracellular pH, cell proliferation and adhesion, and the transfer of CO2 across the renal tubules. It is regulated by hypoxia-inducible factor (HIF) and overexpressed in the setting of tissue hypoxia and acidic tumor microenvironment [54]. Unlike other members of its family, CAIX is suppressed in healthy kidneys by the wild-type von Hippel-Lindau (VHL) protein; hence, normal renal cells are CAIX-negative. Conversely, CAIX is expressed in the vast majority of CRCCs through ubiquination-independent HIF-1α and 2α accumulation due to hypoxia or inactivating mutations in the VHL tumor suppressor gene [55,56].
CAIX strong and diffuse staining is a constant finding in clear cell renal tumors, namely CCRCC and CCRPT. In keeping with that, most tumors with clear cell morphology from a series of 300 unclassified RCC were CAIX-positive [30]. Recently, Baniak et al. confirmed its role as a highly sensitive diagnostic marker in CCRCC and CCPT, while highlighting its low specificity across the whole spectrum of renal epithelial neoplasms (except for RO and ESC-RCC) [57].
A variable degree of CAIX staining can be seen in several nonrenal tumors, including neuroendocrine tumors, mesotheliomas, and tumors of the endometrium, stomach, cervix, breast, lung, liver, testis, and brain, which is a pitfall in distinguishing renal versus nonrenal carcinomas [3].
Viable cells adjacent at the edge of tumor necrosis due to ischemia and hypoxia show non-specific CAIX expression [58].
Cytokeratins (CKs) are intermediate filaments that allow for the identification of epithelial cells; CKs of different molecular weight are characteristic of different epithelial types. In renal tumors, the most common CK is CK7, a low-molecular-weight CK which is highly expressed in most papillary renal cell carcinomas (PRCCs), ChrRCCs, collecting duct carcinomas (CDCs), and almost all CPRCTs and mucinous tubular and spindle cell carcinomas (MTSCCs) [59]. Conversely, CK7 staining is a rare and focal finding in CCRCC and tubulocystic RCC, and it is very focal to almost absent in RO [3]. CK7 is also a marker of urothelial carcinoma; therefore, an upper tract urothelial carcinoma should be always taken into account when assessing a CK7-positive renal neoplasm.
Urothelial carcinoma cells express other CKs, namely CK20 and 34βE12 as well, which are low-molecular-weight and high-molecular-weight CKs, respectively. However, CK20 expression is lacking in the vast majority of renal cell tumors, except ESC-RCC, whereas 34βE12 is a marker of CDC, renal medullary carcinoma (RMC), and most CCPRCTs [3].
CD10 (also known as acute lymphocytic leukemia antigen or CALLA) is a cell-surface glycoprotein expressed on the brush border of the proximal renal tubular epithelial cells and podocytes. It has been traditionally regarded as a marker of CRCC, PRCC, and CDC [3]. Other renal tumors can at least partly express CD10, such as ChrRCC and RO [56]. In the study by Akgul et al. on a series of unclassified RCCs, CD10 was one of the most common markers expressed by tumors with both clear cell RCC-like (92%) and papillary RCC-like/collecting duct carcinoma-like/pure sarcomatoid morphologies (82%) [30]. It is an overall highly sensitive yet non-specific marker for renal tumors, in that its expression can be found in several nonrenal tumors, including carcinomas of the lung, colon, pancreas, ovary, and bladder, as well as soft tissue tumors, melanomas, and lymphomas [60].
CD117 (C-KIT or KIT) is a transmembrane tyrosine kinase receptor encoded by a proto-oncogene that, upon binding to its ligands, regulates cell survival, proliferation, and differentiation.
Several tissues throughout the body show CD117 expression, including hematopoietic precursors, mast cells, and melanocytes, as well as tumors arising from these cells [56].
The combined application of CK7 and CD117 is a reliable first-level panel to discriminate lesions in the spectrum of eosinophilic/oncocytic tumors, with RO being CK7neg/CD117pos, ChrRCC CK7pos/CD117pos, the EVT CK7neg/CD117pos, the LOT CK7pos/CD117neg, and ESC-RCC CK7neg/CD117neg [59] (see Table 3). Sarcomatoid RCC, angiomyolipoma, and pelvic urothelial carcinoma may variably stain for CD117 [54].
Tumors expressing CD117 are susceptible to therapy with tyrosine kinase inhibitors (TKIs), such as imatinib mesylate, which is currently the basis for treatment of gastrointestinal stromal tumors (GISTs), due to the presence of underlying mutations in exons 9 and 11 of the C-KIT gene; however, such mutations have not been identified in CD117-positive renal tumors, thus undermining the efficacy of TKIs in this setting [54,56].
Cathepsin K, a member of the papain-like cysteine peptidase family, is primarily involved in bone resorption but has broader implications in various physiological processes and diseases, including cancer, since its expression and activity are linked to tumor growth, metastasis, and cancer cell invasion [61]. Though not a marker of common histotypes of renal epithelial tumors, it is frequently detected in a subset of renal tumors, as well as in perivascular epithelioid cell tumor (PEComa)/AML [59]. Specifically, among RCCs with a clear cell and papillary architecture, cathepsin K is essential for distinguishing translocation RCC from the more common CCPRCT, which typically lacks expression of this marker. Among renal tumors characterized by eosinophilic cell morphology, cathepsin K expression is observed in ESC-RCC and translocation RCC [3]. Conversely, common eosinophilic/oncocytic renal tumors such as RO, the ChrRCC eosinophilic variant, and oncocytic PRCC are typically negative for cathepsin K, although some variability has been observed in recent studies [62]. Notably, the capillaries and associated macrophages are cathepsin K-positive, thus serving as an internal control for evaluating staining quality.
α-Methylacyl coenzyme A racemase (AMACR) is a mitochondrial enzyme which mediates fatty acid oxidation, and is typically present in normal hepatocytes, proximal renal tubules, and bronchial cells. AMACR is far from being a lineage specific marker: prostatic tumor cells commonly express this AMACR, as well as adenocarcinomas arising from the liver, bladder, colon, ovary, breast [60]. The vast majority of PRCCs are AMACR-positive, along with MTSCRCC and translocation RCC. AMACR expression has been described in up to 78% of CCRCCs according to a recent study, mostly high-grade [63]. Conventional chromophobe/oncocytic renal neoplasms lack this marker.
GATA3 (GATA binding protein 3 to DNA sequence [A/T] GATA[A/G]) is a transcription factor which plays a critical role in stimulating cell proliferation, development, and differentiation. It is regarded as a sensitive, though not specific, marker of urothelial and mammary lineage, in that its expression has been described, at least partly, in several tumor types [3,53]. In the spectrum of renal epithelial tumors, GATA3 is usually expressed by PRNRP and CCPRCT, while being consistently negative in other tumor types.

6. Conclusions

The evolving spectrum of renal tumors, including novel and emerging entities according to the current WHO classification, poses challenges in everyday diagnostic practice. While viewing hematoxylin and eosin-stained slides is still the basis to reliably identify these lesions, IHC strengthens its role as a valuable tool for accurate diagnosis, especially when managing small biopsy specimens; a panel of IHC markers is often needed by the pathologist to support and refine morphological evaluation. In this setting, the pathologist should acknowledge issues and pitfalls of IHC assessment, as well as the specific features of each marker in the proper histopathological context.
The ongoing changes in renal tumor classification are constantly driven by advances in molecular pathology and immunohistochemical techniques, resulting in the definition of entities with more and more precise clinical and prognostic characteristics. The integration of various methodologies and the ongoing search for biomarkers with increasing specificity and sensitivity for individual diagnostic entities are of crucial importance for the accurate management of patients.

Author Contributions

F.S. was involved in conceptualization, methodology, and writing—original draft; R.M. was involved in methodology and writing—original draft; U.G.F. was involved in investigation and formal analysis; A.C. was involved in investigation and formal analysis; M.Z. (Magda Zanelli) was involved in methodology and validation; A.P. was involved in methodology and validation; M.Z. (Maurizio Zizzo) was involved in validation; A.E. was involved in validation; M.B. was involved in validation; A.B.G. was involved in validation; G.C. was involved in supervision and writing—review and editing; and L.C. was involved in writing—original draft and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AMACRα-Methylacyl coenzyme A racemase
CAIXcarbonic anhydrase IX
CCPRCTclear cell papillary renal cell tumor
ChrRCCchromophobe renal cell carcinoma
CKcytokeratins
ESC-RCCeosinophilic solid and cystic renal cell carcinoma
EVTeosinophilic vacuolated tumor
IHCimmunohistochemistry
ISUPInternational Society of Urological Pathology
LOTlow-grade oncocytic tumor
PRCCpapillary renal cell carcinoma
RCCrenal cell carcinoma
TSCtuberous sclerosis complex
VHLvon Hippel Lindau
WHOWorld Health Organization

References

  1. Moch, H.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Gill, A.J.; Hartmann, A.; Menon, S.; Raspollini, M.R.; Rubin, M.A.; Srigley, J.R.; et al. 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. [Google Scholar] [CrossRef] [PubMed]
  2. Alaghehbandan, R.; Siadat, F.; Trpkov, K. What’s new in the WHO 2022 classification of kidney tumours? Pathologica 2022, 115, 8–22. [Google Scholar] [CrossRef] [PubMed]
  3. Li, J.; Wilkerson, M.L.; Deng, F.M.; Liu, H. The Application and Pitfalls of Immunohistochemical Markers in Challenging Diagnosis of Genitourinary Pathology. Arch. Pathol. Lab. Med. 2024, 148, 13–32. [Google Scholar] [CrossRef] [PubMed]
  4. Tickoo, S.K.; de Peralta-Venturina, M.N.; Harik, L.R.; Worcester, H.D.; Salama, M.E.; Young, A.N.; Moch, H.; Amin, M.B. Spectrum of epithelial neoplasms in end-stage renal disease: An experience from 66 tumor-bearing kidneys with emphasis on histologic patterns distinct from those in sporadic adult renal neoplasia. Am. J. Surg. Pathol. 2006, 30, 141–153. [Google Scholar] [CrossRef]
  5. Gobbo, S.; Eble, J.N.; Grignon, D.J.; Martignoni, G.; MacLennan, G.T.; Shah, R.B.; Zhang, S.; Brunelli, M.; Cheng, L. Clear cell papillary renal cell carcinoma: A distinct histopathologic and molecular genetic entity. Am. J. Surg. Pathol. 2008, 32, 1239–1245. [Google Scholar] [CrossRef] [PubMed]
  6. Williamson, S.R. Clear cell papillary renal cell carcinoma: An update after 15 years. Pathology 2021, 53, 109–119. [Google Scholar] [CrossRef] [PubMed]
  7. Massari, F.; Ciccarese, C.; Hes, O.; Michal, M.; Caliò, A.; Fiorentino, M.; Giunchi, F.; D’Amuri, A.; Sanguedolce, F.; Sabbatini, R.; et al. The Tumor Entity Denominated “clear cell-papillary renal cell carcinoma” According to the WHO 2016 new Classification, have the Clinical Characters of a Renal Cell Adenoma as does Harbor a Benign Outcome. Pathol. Oncol. Res. 2018, 24, 447–456. [Google Scholar] [CrossRef] [PubMed]
  8. Gupta, S.; Inwards, C.Y.; Van Dyke, D.L.; Jimenez, R.E.; Cheville, J.C. Defining clear cell papillary renal cell carcinoma in routine clinical practice. Histopathology 2020, 76, 1093–1095. [Google Scholar] [CrossRef] [PubMed]
  9. Xu, J.; Reznik, E.; Lee, H.J.; Gundem, G.; Jonsson, P.; Sarungbam, J.; Bialik, A.; Sanchez-Vega, F.; Creighton, C.J.; Hoekstra, J.; et al. Abnormal oxidative metabolism in a quiet genomic background underlies clear cell papillary renal cell carcinoma. Elife 2019, 8, e38986. [Google Scholar] [CrossRef]
  10. Mantilla, J.G.; Antic, T.; Tretiakova, M. GATA3 as a valuable marker to distinguish clear cell papillary renal cell carcinomas from morphologic mimics. Hum. Pathol. 2017, 66, 152–158. [Google Scholar] [CrossRef]
  11. Martignoni, G.; Brunelli, M.; Segala, D.; Munari, E.; Gobbo, S.; Cima, L.; Borze, I.; Wirtanen, T.; Sarhadi, V.K.; Atanesyan, L.; et al. Validation of 34betaE12 immunoexpression in clear cell papillary renal cell carcinoma as a sensitive biomarker. Pathology 2017, 49, 10–18. [Google Scholar] [CrossRef] [PubMed]
  12. Williamson, S.R.; Gill, A.J.; Argani, P.; Chen, Y.B.; Egevad, L.; Kristiansen, G.; Grignon, D.J.; Hes, O. Report from the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers: III: Molecular Pathology of Kidney Cancer. Am. J. Surg. Pathol. 2020, 44, e47–e65. [Google Scholar] [CrossRef] [PubMed]
  13. Kim, M.; Joo, J.W.; Lee, S.J.; Cho, Y.A.; Park, C.K.; Cho, N.H. Comprehensive Immunoprofiles of Renal Cell Carcinoma Subtypes. Cancers 2020, 12, 602. [Google Scholar] [CrossRef] [PubMed]
  14. Griffin, B.B.; Lin, X. Cytomorphologic analysis of clear cell papillary renal cell carcinoma: Distinguishing diagnostic features. Cancer Cytopathol. 2021, 129, 192–203. [Google Scholar] [CrossRef] [PubMed]
  15. Akgul, M.; Williamson, S.R. Immunohistochemistry for the diagnosis of renal epithelial neoplasms. Semin. Diagn. Pathol. 2022, 39, 1–16. [Google Scholar] [CrossRef] [PubMed]
  16. Reuter, V.E.; Argani, P.; Zhou, M.; Delahunt, B.; Members of the ISUP Immunohistochemistry in Diagnostic Urologic Pathology Group. Best practices recommendations in the application of immunohistochemistry in the kidney tumors: Report from the International Society of Urologic Pathology consensus conference. Am. J. Surg. Pathol. 2014, 38, e35–e49. [Google Scholar] [CrossRef] [PubMed]
  17. 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. [Google Scholar] [CrossRef] [PubMed]
  18. Argani, P. A Molecular Marker for Eosinophilic Solid and Cystic Renal Cell Carcinoma. Eur. Urol. 2018, 74, 487–488. [Google Scholar] [CrossRef] [PubMed]
  19. Trpkov, K.; Williamson, S.R.; Gill, A.J.; Adeniran, A.J.; Agaimy, A.; Alaghehbandan, R.; Amin, M.B.; Argani, P.; Chen, Y.B.; Cheng, L.; et al. Novel, emerging and provisional renal entities: The Genitourinary Pathology Society (GUPS) update on renal neoplasia. Mod. Pathol. 2021, 34, 1167–1184. [Google Scholar] [CrossRef]
  20. Trpkov, K.; Hes, O.; Agaimy, A.; Bonert, M.; Martinek, P.; Magi-Galluzzi, C.; Kristiansen, G.; Lüders, C.; Nesi, G.; Compérat, E.; et al. Fumarate hydratase-deficient renal cell carcinoma is strongly correlated with fumarate hydratase mutation and hereditary Leiomyomatosis and renal cell carcinoma syndrome. Am. J. Surg. Pathol. 2016, 40, 865–875. [Google Scholar] [CrossRef]
  21. Guo, J.; Tretiakova, M.S.; Troxell, M.L.; Osunkoya, A.O.; Fadare, O.; Sangoi, A.R.; Shen, S.S.; Lopez-Beltran, A.; Mehra, R.; Heider, A.; et al. Tuberous sclerosis-associated renal cell carcinoma: A clinicopathologic study of 57 separate carcinomas in 18 patients. Am. J. Surg. Pathol. 2014, 38, 1457–1467. [Google Scholar] [CrossRef] [PubMed]
  22. Schreiner, A.; Daneshmand, S.; Bayne, A.; Countryman, G.; Corless, C.L.; Troxell, M.L. Distinctive morphology of renal cell carcinomas in tuberous sclerosis. Int. J. Surg. Pathol. 2010, 18, 409–418. [Google Scholar] [CrossRef] [PubMed]
  23. Palsgrove, D.N.; Li, Y.; Pratilas, C.A.; Lin, M.T.; Pallavajjalla, A.; Gocke, C.; De Marzo, A.M.; Matoso, A.; Netto, G.J.; Epstein, J.I.; et al. Eosinophilic solid and cystic (ESC) renal cell carcinomas harbor TSC mutations: Molecular analysis supports an expanding clinicopathologic spectrum. Am. J. Surg. Pathol. 2018, 42, 1166–1181. [Google Scholar] [CrossRef] [PubMed]
  24. McKenney, J.K.; Przybycin, C.G.; Trpkov, K.; Magi-Galluzzi, C. Eosinophilic solid and cystic renal cell carcinomas have metastatic potential. Histopathology 2018, 72, 1066–1067. [Google Scholar] [CrossRef] [PubMed]
  25. Tretiakova, M.S. Eosinophilic solid and cystic renal cell carcinoma mimicking epithelioid angiomyolipoma: Series of 4 primary tumors and 2 metastases. Hum. Pathol. 2018, 80, 65–75. [Google Scholar] [CrossRef] [PubMed]
  26. Trpkov, K.; Abou-Ouf, H.; Hes, O.; Lopez, J.I.; Nesi, G.; Comperat, E.; Sibony, M.; Osunkoya, A.O.; Zhou, M.; Gokden, N.; et al. Eosinophilic Solid and Cystic Renal Cell Carcinoma (ESC RCC): Further Morphologic and Molecular Characterization of ESC RCC as a Distinct Entity. Am. J. Surg. Pathol. 2017, 41, 1299–1308. [Google Scholar] [CrossRef] [PubMed]
  27. Lobo, J.; Ohashi, R.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Cree, I.A.; Gill, A.J.; Hartmann, A.; Menon, S.; Netto, G.J.; et al. WHO 2022 landscape of papillary and chromophobe renal cell carcinoma. Histopathology 2022, 81, 426–438. [Google Scholar] [CrossRef] [PubMed]
  28. Siadat, F.; Trpkov, K. ESC, ALK, HOT and LOT: Three Letter Acronyms of Emerging Renal Entities Knocking on the Door of the WHO Classification. Cancers 2020, 12, 168. [Google Scholar] [CrossRef]
  29. Li, Y.; Reuter, V.E.; Matoso, A.; Netto, G.J.; Epstein, J.I.; Argani, P. Re-evaluation of 33 ‘unclassified’ eosinophilic renal cell carcinomas in young patients. Histopathology 2018, 72, 588–600. [Google Scholar] [CrossRef]
  30. Akgul, M.; Cheng, L. Immunophenotypic and pathologic heterogeneity of unclassified renal cell carcinoma: A study of 300 cases. Hum. Pathol. 2020, 102, 70–78. [Google Scholar] [CrossRef]
  31. Pivovarcikovam, K.; Alaghehbandan, R.; Vanecek, T.; Ohashi, R.; Pitra, T.; Hes, O. TSC/ mTOR pathway mutation associated eosinophilic/oncocytic renal neoplasms: A heterogeneous group of tumors with distinct morphology, immunohistochemical profile, and similar genetic background. Biomedicine 2022, 10, 322. [Google Scholar]
  32. Hartmann, A.; Agaimy, A. Eosinophiles solide und zystisches Nierenzellkarzinom (ESC-NZK) [Eosinophilic, solid, and cystic renal cell carcinoma (ESC-RCC)]. Pathologe 2021, 42, 565–570, Erratum in Pathologe 2021, Online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  33. Caliò, A.; Marletta, S.; Brunelli, M.; Martignoni, G. WHO 2022 Classification of Kidney Tumors: What is relevant? An update and future novelties for the pathologist. Pathologica 2022, 115, 23–31. [Google Scholar] [CrossRef] [PubMed]
  34. Mansoor, M.; Siadat, F.; Trpkov, K. Low-grade oncocytic tumor (LOT)—A new renal entity ready for a prime time: An updated review. Histol. Histopathol. 2022, 37, 405–413. [Google Scholar]
  35. He, H.; Trpkov, K.; Martinek, P.; Isikci, O.T.; Maggi-Galuzzi, C.; Alaghehbandan, R.; Gill, A.J.; Tretiakova, M.; Lopez, J.I.; Williamson, S.R.; et al. “High-grade oncocytic renal tumor”: Morphologic, immunohistochemical, and molecular genetic study of 14 cases. Virchows Arch. 2018, 473, 725–738. [Google Scholar] [CrossRef] [PubMed]
  36. Chen, Y.-B.; Mirsadraei, L.; Jayakumaran, G.; Al-Ahmadie, H.A.; Fine, S.W.; Gopalan, A.; Sirintrapun, S.J.; Tickoo, S.K.; Reuter, V.E. Somatic mutations of TSC2 or MTOR characterize a morphologically distinct subset of sporadic renal cell carcinoma with eosinophilic and vacuolated cytoplasm. Am. J. Surg. Pathol. 2019, 43, 121–131. [Google Scholar] [CrossRef] [PubMed]
  37. Xia, Q.Y.; Wang, X.T.; Zhao, M.; He, H.Y.; Fang, R.; Ye, S.B.; Li, R.; Wang, X.; Zhang, R.S.; Lu, Z.F.; et al. TSC/MTOR-associated eosinophilic renal tumors exhibit a heterogeneous clinicopathologic spectrum: A targeted next-generation sequencing and gene expression profiling study. Am. J. Surg. Pathol. 2022, 46, 1562–1576. [Google Scholar] [CrossRef] [PubMed]
  38. Farcaş, M.; Gatalica, Z.; Trpkov, K.; Swensen, J.; Zhou, M.; Alaghehbandan, R.; Williamson, S.R.; Magi-Galluzzi, C.; Gill, A.J.; Tretiakova, M.; et al. Eosinophilic vacuolated tumor (EVT) of kidney demonstrates sporadic TSC/MTOR mutations: Next-generation sequencing multi-institutional study of 19 cases. Mod. Pathol. 2022, 35, 344–351. [Google Scholar] [CrossRef] [PubMed]
  39. Lerma, L.A.; Schade, G.R.; Tretiakova, M.S. Co-existence of ESC-RCC, EVT, and LOT as synchronous and metachronous tumors in six patients with multifocal neoplasia but without clinical features of tuberous sclerosis complex. Hum. Pathol. 2021, 116, 1–11. [Google Scholar] [CrossRef] [PubMed]
  40. Kapur, P.; Gao, M.; Zhong, H.; Rakheja, D.; Cai, Q.; Pedrosa, I.; Margulis, V.; Xu, L.; Kinch, L.; Brugarolas, J. Eosinophilic vacuolated tumor of the kidney: A review of evolving concepts in this novel subtype with additional insights from a case with MTOR mutation and concomitant chromosome 1 loss. Adv. Anat. Pathol. 2021, 28, 251–257. [Google Scholar] [CrossRef]
  41. Hes, O.; Trpkov, K. 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. [Google Scholar] [CrossRef] [PubMed]
  42. Trpkov, K.; Williamson, S.R.; Gao, Y.; Martinek, P.; Cheng, L.; Sangoi, A.R.; Yilmaz, A.; Wang, C.; Fraile, P.S.M.; Montiel, D.M.P.; et al. Low-grade oncocytic tumor of kidney (CD117 Negative, Cytokeratin 7 Positive): A distinct entity? Histopathology 2019, 75, 174–184. [Google Scholar] [CrossRef] [PubMed]
  43. Kravtsov, O.; Gupta, S.; Cheville, J.C.; Sukov, W.R.; Rowsey, R.; Herrera-Hernandez, L.P.; Lohse, C.M.; Knudson, R.; Leibovich, B.C.; Jimenez, R.E. Low-grade oncocytic tumor of kidney (CK7-positive, CD117-negative): Incidence in a single institutional experience with clinicopathological and molecular characteristics. Hum. Pathol. 2021, 114, 9–18. [Google Scholar] [CrossRef] [PubMed]
  44. Kapur, P.; Gao, M.; Zhong, H.; Chintalapati, S.; Mitui, M.; Barnes, S.D.; Zhou, Q.; Miyata, J.; Carrillo, D.; Malladi, V.S.; et al. Germline and sporadic mTOR pathway mutations in low-grade oncocytic tumor of the kidney. Mod. Pathol. 2022, 35, 333–343. [Google Scholar] [CrossRef]
  45. Morini, A.; Drossart, T.; Timsit, M.-O.; Sibony, M.; Vasiliu, V.; Gimenez-Roqueplo, A.-P.; Favier, J.; Badoual, C.; Mejean, A.; Burnichon, N.; et al. Low-grade oncocytic renal tumor (LOT): Mutations in mTOR pathway genes and low expression of FOXI1. Mod. Pathol. 2022, 35, 352–360. [Google Scholar] [CrossRef] [PubMed]
  46. Akgul, M.; Al-Obaidy, I.K.; Cheng, L.; Idrees, M.T. Low-grade oncocytic tumour expands the spectrum of renal oncocytic tumours and deserves separate classification: A review of 23 cases from a single tertiary institute. J. Clin. Pathol. 2021, 75, 772–775. [Google Scholar] [CrossRef] [PubMed]
  47. Williamson, S.R.; Hes, O.; Trpkov, K.; Aggarwal, A.; Satapathy, A.; Mishra, S.; Sharma, S.; Sangoi, A.; Cheng, L.; Akgul, M.; et al. Low-grade oncocytic tumour of the kidney is characterised by genetic alterations of TSC1, TSC2, MTOR or PIK3CA and consistent GATA3 positivity. Histopathology 2023, 82, 296–304. [Google Scholar] [CrossRef] [PubMed]
  48. Gupta, S.; Rowsey, R.A.; Cheville, J.C.; Jimenez, R.E. Morphologic overlap between low-grade oncocytic tumor and eosinophilic variant of chromophobe renal cell carcinoma. Hum. Pathol. 2022, 119, 114–116. [Google Scholar] [CrossRef] [PubMed]
  49. Athanazio, D.A.; Amorim, L.S.; da Cunha, I.W.; Leite, K.R.M.; da Paz, A.R.; Gomes, R.d.P.X.; Tavora, F.R.F.; Faraj, S.F.; Cavalcanti, M.S.; Bezerra, S.M. Classification of renal cell tumors—Current concepts and use of ancillary tests: Recommendations of the Brazilian Society of Pathology. Surg. Exp. Pathol. 2021, 4, 4. [Google Scholar] [CrossRef]
  50. Mohanty, S.K.; Satapathy, A.; Aggarwal, A.; Mishra, S.K.; Sampat, N.Y.; Sharma, S.; Williamson, S. Oncocytic renal neoplasms with diffuse keratin 7 immunohistochemistry harbor frequent alterations in the mammalian target of rapamycin pathway. Mod. Pathol. 2022, 35, 361–375. [Google Scholar] [CrossRef]
  51. Hes, O.; Michal, M.; Kuroda, N.; Martignoni, G.; Brunelli, M.; Lu, Y.; Adley, B.P.; Alvarado-Cabrero, I.; Yang, X.J. Vimentin reactivity in renal oncocytoma: Immunohistochemical study of 234 cases. Arch. Pathol. Lab. Med. 2007, 131, 1782–1788. [Google Scholar] [CrossRef]
  52. Ricci, C.; Ambrosi, F.; Franceschini, T.; Giunchi, F.; Grillini, A.; Franchini, E.; Grillini, M.; Schiavina, R.; Massari, F.; Mollica, V.; et al. Evaluation of an institutional series of low-grade oncocytic tumor (LOT) of the kidney review of the mutational landscape of LOT. Virchows Arch. 2023, 483, 687–698. [Google Scholar] [CrossRef] [PubMed]
  53. Miettinen, M.; McCue, P.; Sarlomo-Rikala, M.; Rys, J.; Czapiewski, P.; Wazny, K.; Langfort, R.; Waloszczyk, P.; Biernat, W.; Lasota, J.; et al. GATA3: A multispecific but potentially useful marker in surgical pathology: A systematic analysis of 2500 epithelial nonepithelial tumors. Am. J. Surg. Pathol. 2014, 38, 13–22. [Google Scholar] [CrossRef]
  54. Truong, L.D.; Shen, S.S. Immunohistochemical diagnosis of renal neoplasms. Arch. Pathol. Lab. Med. 2011, 135, 92–109. [Google Scholar] [CrossRef]
  55. Tuffaha, M.S.A.; Guski, H.; Kristiansen, G. Markers and Immunoprofile of Renal and Urinary Tract Tumors. In Immunohistochemistry in Tumor Diagnostics; Springer: Cham, Switzerland, 2018. [Google Scholar] [CrossRef]
  56. Netto, G.J.; Epstein, J.I. Immunohistology of the Bladder, Kidney, and Testis. In Diagnostic Immunohistochemistry Theranostic and Genomic Applications, 5th ed.; Dabbs, D.J., Ed.; Elsevier: Amsterdam, The Netherlands, 2019. [Google Scholar]
  57. Baniak, N.; Flood, T.A.; Buchanan, M.; Dal Cin, P.; Hirsch, M.S. Carbonic anhydrase IX (CA9) expression in multiple renal epithelial tumour subtypes. Histopathology 2020, 77, 659–666. [Google Scholar] [CrossRef] [PubMed]
  58. Dorai, T.; Sawczuk, I.S.; Pastorek, J.; Wiernik, P.H.; Dutcher, J.P. The role of carbonic anhydrase IX overexpression in kidney cancer. Eur. J. Cancer 2005, 41, 2935–2947. [Google Scholar] [CrossRef]
  59. Farber, N.J.; Kim, C.J.; Modi, P.K.; Hon, J.D.; Sadimin, E.T.; Singer, E.A. Renal cell carcinoma: The search for a reliable biomarker. Transl. Cancer Res. 2017, 6, 620–632. [Google Scholar] [CrossRef] [PubMed]
  60. Shen, S.S.; Truong, L.D.; Scarpelli, M.; Lopez-Beltran, A. Role of immunohistochemistry in diagnosing renal neoplasms: When is it really useful? Arch. Pathol. Lab. Med. 2012, 136, 410–417. [Google Scholar] [CrossRef]
  61. Caliò, A.; Brunelli, M.; Gobbo, S.; Argani, P.; Munari, E.; Netto, G.; Martignoni, G. Cathepsin K: A Novel Diagnostic and Predictive Biomarker for Renal Tumors. Cancers 2021, 13, 2441. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Iakymenko, O.A.; Delma, K.S.; Jorda, M.; Kryvenko, O.N. Cathepsin K (Clone EPR19992) Demonstrates Uniformly Positive Immunoreactivity in Renal Oncocytoma, Chromophobe Renal Cell Carcinoma, and Distal Tubules. Int. J. Surg. Pathol. 2021, 29, 600–605. [Google Scholar] [CrossRef]
  63. Rotterova, P.; Alaghehbandan, R.; Skopal, J.; Rogala, J.; Slisarenko, M.; Strakova Peterikova, A.; Michalova, K.; Montiel, D.P.; Farcas, M.; Ulamec, M.; et al. Alpha-methyl CoA racemase (AMACR) reactivity across the spectrum of clear cell renal cell neoplasms. Ann. Diagn. Pathol. 2024, 71, 152297. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clear cell papillary renal cell tumor. (A,B) Hematoxylin–eosin (H&E)-stained section showing tumor cells with characteristic suprabasal nuclei, arranged in branched, sometimes cystic, tubular formations (original magnifications ×100 (A) and ×200 (B)). (C,D) Tumor cells are positive for CK7 (original magnification ×100 (C)), and CAIX, with the typical cup-shaped staining pattern (original magnification ×200 (D)). (E) Tumor cells are negative for AMACR (original magnification ×100).
Figure 1. Clear cell papillary renal cell tumor. (A,B) Hematoxylin–eosin (H&E)-stained section showing tumor cells with characteristic suprabasal nuclei, arranged in branched, sometimes cystic, tubular formations (original magnifications ×100 (A) and ×200 (B)). (C,D) Tumor cells are positive for CK7 (original magnification ×100 (C)), and CAIX, with the typical cup-shaped staining pattern (original magnification ×200 (D)). (E) Tumor cells are negative for AMACR (original magnification ×100).
Cancers 16 01856 g001
Figure 2. Low-grade oncocytic tumor. (A) Hematoxylin–eosin (H&E)-stained section showing eosinophilic tumor cells, often with nuclear halos (inset) (original magnifications ×100 and ×200 (inset)). (B,C) Tumor cells are diffusely positive for PAX8 ((B) original magnification ×100), and CK7 ((C) original magnification ×100). (D) Tumor cells are negative for CD117 (original magnification ×100); scattered positive mast cells.
Figure 2. Low-grade oncocytic tumor. (A) Hematoxylin–eosin (H&E)-stained section showing eosinophilic tumor cells, often with nuclear halos (inset) (original magnifications ×100 and ×200 (inset)). (B,C) Tumor cells are diffusely positive for PAX8 ((B) original magnification ×100), and CK7 ((C) original magnification ×100). (D) Tumor cells are negative for CD117 (original magnification ×100); scattered positive mast cells.
Cancers 16 01856 g002
Table 1. Brief summary of the changes in the fifth edition of the WHO classification of renal tumors which are discussed in this review, along with the underlying reason.
Table 1. Brief summary of the changes in the fifth edition of the WHO classification of renal tumors which are discussed in this review, along with the underlying reason.
WHO 2022WHO 2016Reason
Clear cell papillary renal cell tumor (CCPRCT)Clear cell papillary renal cell carcinoma (CCPRCC)Benign behavior
Eosinophilic solid and cystic renal cell carcinoma (ESC-RCC)Not presentNew entity in the subgroup of “Other renal tumours”
Low-grade oncocytic tumor (LOT)Not presentEmerging entity in the subgroup of “Other oncocytic tumours of the kidney”
Eosinophilic vacuolated tumor (EVT)Not presentEmerging entity in the subgroup of “Other oncocytic tumours of the kidney”
Table 2. Differential diagnosis of low-grade tumors with clear cell and/or papillary features. CCPRCT: clear cell papillary renal cell tumor; CCRCC: clear cell renal cell carcinoma; PRCC: papillary renal cell carcinoma.
Table 2. Differential diagnosis of low-grade tumors with clear cell and/or papillary features. CCPRCT: clear cell papillary renal cell tumor; CCRCC: clear cell renal cell carcinoma; PRCC: papillary renal cell carcinoma.
MarkersCCRCC CCPRCT PRCC
CAIX+Cancers 16 01856 i001+Cancers 16 01856 i002
CK7−/+Cancers 16 01856 i003+Cancers 16 01856 i001+
AMACR−/+Cancers 16 01856 i001−/+Cancers 16 01856 i002+
CD10+Cancers 16 01856 i002−/+Cancers 16 01856 i002+
34βE12Cancers 16 01856 i002+/−Cancers 16 01856 i002
GATA3Cancers 16 01856 i002+Cancers 16 01856 i002
Table 3. Immunohistochemical markers in selected epithelial oncocytic tumors.
Table 3. Immunohistochemical markers in selected epithelial oncocytic tumors.
CK7CD117Cathepsin KCK20
RO+
ChrRCC, eos++
EVT−/+++
LOT+
ESC-RCC++
ChrRCC, eos: chromophobe renal cell carcinoma, eosinophilic variant; ESC-RCC: eosinophilic solid and cystic renal cell carcinoma; EVT: eosinophilic vacuolated tumor; LOT: low-grade oncocytic tumor; RO: renal oncocytoma.
Table 4. Diagnostic markers in different types of renal tumors.
Table 4. Diagnostic markers in different types of renal tumors.
BiomarkerStaining PatternMain Expression
CAIXmembranous/cytoplasmicCCRCC, CCPRCT
CK7cytoplasmicPRCC, ChrRCC, CDC, CCRPCT, MTSCC, LOT
CK20cytoplasmicESC-RCC
34βE12cytoplasmicCDC, RMC, CCPRCT
CD10membranousCRCC, PRCC, CDC
CD117membranous/cytoplasmicChrRCC, RO, EVT
cathepsin KcytoplasmicEVT, ESC-RCC, AML
AML: angiomyolipoma; CCPRCT: clear cell papillary renal cell tumor; CCRCC: clear cell renal cell carcinoma; ChrRCC: chromophobe renal cell carcinoma; CDC: collecting duct carcinoma; ESC-RCC: eosinophilic solid and cystic renal cell carcinoma; EVT: eosinophilic vacuolate tumor; LOT: low-grade oncocytic tumor; MTSCC: mucinous tubular and spindle cell carcinoma; PRCC: papillary renal cell carcinoma; RMC: renal medullary carcinoma; RO: renal oncocytoma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sanguedolce, F.; Mazzucchelli, R.; Falagario, U.G.; Cormio, A.; Zanelli, M.; Palicelli, A.; Zizzo, M.; Eccher, A.; Brunelli, M.; Galosi, A.B.; et al. Diagnostic Biomarkers in Renal Cell Tumors According to the Latest WHO Classification: A Focus on Selected New Entities. Cancers 2024, 16, 1856. https://doi.org/10.3390/cancers16101856

AMA Style

Sanguedolce F, Mazzucchelli R, Falagario UG, Cormio A, Zanelli M, Palicelli A, Zizzo M, Eccher A, Brunelli M, Galosi AB, et al. Diagnostic Biomarkers in Renal Cell Tumors According to the Latest WHO Classification: A Focus on Selected New Entities. Cancers. 2024; 16(10):1856. https://doi.org/10.3390/cancers16101856

Chicago/Turabian Style

Sanguedolce, Francesca, Roberta Mazzucchelli, Ugo Giovanni Falagario, Angelo Cormio, Magda Zanelli, Andrea Palicelli, Maurizio Zizzo, Albino Eccher, Matteo Brunelli, Andrea Benedetto Galosi, and et al. 2024. "Diagnostic Biomarkers in Renal Cell Tumors According to the Latest WHO Classification: A Focus on Selected New Entities" Cancers 16, no. 10: 1856. https://doi.org/10.3390/cancers16101856

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop